WO2007100650A2 - Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway - Google Patents

Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway Download PDF

Info

Publication number
WO2007100650A2
WO2007100650A2 PCT/US2007/004707 US2007004707W WO2007100650A2 WO 2007100650 A2 WO2007100650 A2 WO 2007100650A2 US 2007004707 W US2007004707 W US 2007004707W WO 2007100650 A2 WO2007100650 A2 WO 2007100650A2
Authority
WO
WIPO (PCT)
Prior art keywords
myd88
cancer
cancer cell
tlr4
cell
Prior art date
Application number
PCT/US2007/004707
Other languages
French (fr)
Other versions
WO2007100650A3 (en
Inventor
Guillermo G. Mor
Original Assignee
Yale University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Yale University filed Critical Yale University
Priority to US12/224,244 priority Critical patent/US7985538B2/en
Priority to EP07751467A priority patent/EP1996935B1/en
Priority to AT07751467T priority patent/ATE524732T1/en
Publication of WO2007100650A2 publication Critical patent/WO2007100650A2/en
Publication of WO2007100650A3 publication Critical patent/WO2007100650A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/44Multiple drug resistance

Definitions

  • ovarian cancer is not diagnosed until it has progressed and the prognosis is not good.
  • the standard first-line chemotherapy for patients with ovarian cancer is a platinum-taxane combination regimen. More than eighty percent of patients will respond initially to this reatement. However, fewer than 10% will remain in remission.
  • a cellular marker, MyD88 useful for assessing an individual's (patient's) sensitivity (or resistance) to chemotherapy, particularly sensitivity (or resistance) to chemotherapeutic drugs, such as plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel).
  • chemotherapeutic drugs such as plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel).
  • plant alkaloids e.g., a taxane, such as paclitaxel or docetaxel.
  • TLRs Toll-like receptors
  • TLR-4 signaling a link between TLR-4-MyD88 signaling, inflammation, tumor growth and chemoresistance.
  • Described herein is the expression of TLR-4 in cancer cells, as exemplified by epithelial ovarian cancer (EOC) cells and the differential effect of TLR-4 ligation by LPS and paclitaxel in MyD88 positive (MyD88 + ) and MyD88 negative (MyD88 " ) EOC cells.
  • EOC epithelial ovarian cancer
  • the work described herein provides the basis for assessing responsiveness (sensitivity or resistance) to chemotherapeutic drugs, such as a woman's responsiveness (sensitivity or resistance) to chemotherapeutic drugs, particularly paclitaxel, as well as for treating or reducing the development of a cancer characterized by elevated expression of MyD88.
  • EOC cells demonstrate that in EOC cells, signaling through TLR-4 and MyD88 represent a major source of pro-inflammatory cytokines, which promote tumor growth. Furthermore, they have identified a new mechanism for the acquisition of paclitaxel chemo-resistance and therefore, a novel target for the development of molecularly directed therapy in paclitaxel resistant ovarian cancer.
  • EOC cells that express MyD88 show an increase in cell proliferation, secretion and production of proinflammatory cytokines and activation of anti-apoptotic proteins.
  • Cells that do not express MyD88 are paclitaxel sensitive, do not shown NF- ⁇ B activation and do not produce inflammatory cytokines.
  • this invention relates to a method of assessing the sensitivity or resistance of cancer cells to chemotherapy, particularly chemotherapy with a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel), comprising assaying MyD88 expression in the cancer cells.
  • MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself.
  • nucleic acid encoding MyD88 e.g., mRNA, DNA
  • MyD88 expression indicates that the individual (patient) from whom the sample was obtained is resistant to such chemotherapy (e.g., chemotherapy with a plant alkaloid, such as a taxane, such as paclitaxel.or docetaxel).
  • chemotherapy e.g., chemotherapy with a plant alkaloid, such as a taxane, such as paclitaxel.or docetaxel.
  • the sample is negative for MyD88 expression, the individual (patient) is sensitive to such chemotherapy.
  • cancer cells e.g., ovarian cancer cells
  • MyD88 ⁇ they are sensitive to chemotherapy with a plant alkaloid (e.g., a taxane, such as paclitaxel or docetaxel).
  • MyD88 positive cancer cells e.g., ovarian cancer cells
  • a plant alkaloid e.g., a taxane, such as paclitaxel or docetaxel
  • the level of MyD88 in cancer cells e.g.
  • a reference or standard e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity /resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel.
  • a reference or standard e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity /resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel.
  • the relationship between MyD88 level in a type of cancer cell e.g., ovarian cancer cells
  • resistance to a plant alkaloid such as a taxane (e.g., paclitaxel or docetaxel)
  • a taxane e.g., pac
  • Resistance/sensitivity to such a chemotherapeutic can be correlated with a range of MyD88 levels and a scale produced.
  • MyD88 level determined in a sample obtained from an individual e.g., MyD88 level in ovarian cancer cells
  • an indication of resistance or sensitivity determined e.g., a "cutoff' MyD88 value (e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel))
  • a "cutoff' MyD88 value e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel)
  • levels determined for cells obtained from a patient compared with that value, to assess their resistance or sensitivity to the plant alkaloid.
  • the cancer cells are resistant to such chemotherapy. If the level of MyD 88 is less than the cutoff value, the cells are sensitive to such chemotherapy.
  • the method can be carried out to assess the sensitivity or resistance of an individual suffering from, for example, ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
  • the invention relates to a method of assessing the sensitivity or resistance of cancer cells to a taxane, such as paclitaxel or docetaxel, used to treat cancer, such as ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
  • the method of assessing the sensitivity or resistance of cancer cells to chemotherapy comprises assaying MyD88 expression in the cancer cells.
  • MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself.
  • nucleic acid encoding MyD88 (e.g., mRNA, DNA) can be assessed.
  • MyD88 expression indicates that the individual (patient) from whom the sample was obtained is resistant to such chemotherapy (e.g., chemotherapy with a taxane, such as paclitaxel. or docetaxel).
  • chemotherapy e.g., chemotherapy with a taxane, such as paclitaxel. or docetaxel.
  • the level of MyD88 in cancer cells e.g.
  • a reference or standard e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity/resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel.
  • a reference or standard e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity/resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel.
  • the relationship between MyD88 level in a type of cancer cell e.g., ovarian cancer cells
  • resistance to a taxane e.g., paclitaxel or docetaxel
  • Resistance/sensitivity to a taxane can be correlated with a range of MyD88 levels and a scale produced.
  • MyD88 level determined in a sample obtained from an individual e.g., MyD88 level in ovarian cancer cells
  • the reference expression level can be, for example, the level of MyD88 in noncancer cells, such as ovarian cells that are not cancer cells or other cells (e.g., cells from a source other than ovarian tissue), which may or may not be cancer cells.
  • reference cells can be obtained from the individual whose paclitaxel sensitivity /resistance is being assessed or from another individual or individuals.
  • the reference expression levels can be determined at the time an individual is assessed for paclitaxel sensitivity/resistance or can be pre- established reference expression levels (reference expression standard).
  • a "cutoff MyD88 value e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a taxane (e.g., paclitaxel or docetaxel)
  • a "cutoff MyD88 value e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a taxane (e.g., paclitaxel or docetaxel)
  • levels determined for cells obtained from a patient compared with that value, to assess their resistance or sensitivity to the plant alkaloid.
  • the cancer cells are resistant to such chemotherapy. If the level of MyD88 is less than the cutoff value, the cells are sensitive to such chemotherapy.
  • the method can be carried out to assess the sensitivity or resistance of ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, or Kaposi's sarcoma cells.
  • ovarian cancer cells e.g., epithelial ovarian cancer cells
  • breast cancer cells e.g., head and/or neck cancer cells
  • prostate cancer cells e.g., non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, or Kaposi's sarcoma cells.
  • the invention relates to a method of determining MyD88 (e.g., level of expression, presence or absence of expression) in cells isolated from a tumor, such as by using laser microdissection and dot-blot analysis.
  • laser microdissection is used to obtain a tumor sample that contains a sufficient number of cancer cells to permit assessment of MyD88 (e.g. from about 500 to about 5,000 cells).
  • the sample can be obtained, for example, from a biopsy or a tumor after surgery. Frozen sections can be used, for example.
  • the cells are lysed, using known methods and the product is analyzed (e.g., by Western blot) for MyD88 expression.
  • MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself.
  • nucleic acid encoding MyD88 e.g., mRNA, DNA
  • MyD88 expression indicates that the patient from whom the sample was obtained is resistant to paclitaxel. Conversely, if the sample is negative for MyD88 expression, the patient is sensitive to paclitaxel and the patient's prognosis is good (e.g., better than if the patient is resistant to paclitaxel).
  • Samples used for the analysis can be obtained by other methods known to those of skill in the art and can be analyzed for MyD88 expression by other art recognized methods as well.
  • Also described herein is a method of inhibiting proliferation of a cancer cell, comprising contacting the cell (such as an ovarian cancer cell, particularly human ovarian cancer cells) with a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway.
  • this invention relates to a method of reducing production of proinflammatory cytokines, which promote tumor growth, by cancer cells and, as a result, reducing the extent to which tumor growth occurs.
  • Such methods can be carried out to inhibit proliferation of cancer cells of many types in which there is an active TLR4:MyD88 signaling pathway, including, but not limited to, ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
  • ovarian cancer cells e.g., epithelial ovarian cancer cells
  • breast cancer cells e.g., epithelial ovarian cancer cells
  • breast cancer cells e.g., head and/or neck cancer cells
  • prostate cancer cells e.g., non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
  • This invention is also a method of reducing (partially or totally) tumor growth, comprising contacting tumor cells in which there is an active TLR4:MyD88 signaling pathway with a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 signaling pathway.
  • the subject of this invention is a method of treating or preventing cancer, such as a cancer characterized by an active TLR4:MyD88 signaling pathway, in an individual in need thereof, comprising administering to the individual (e.g., a human suffering from or at risk of developing such a cancer) an effective amount of a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway.
  • the invention is a method of treating or preventing ovarian cancer, in an individual (such as a woman suffering from or at risk of developing ovarian cancer characterized by an active TLR4:MyD88 signaling pathway), comprising administering to the individual (e.g., the woman) an effective amount of a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway.
  • a method for inhibiting proliferation of a cancer cell comprising contacting the cell (such as an ovarian cancer cell, particularly human ovarian cancer cells) with a compound that inhibits signaling through the TLR4:MyD88 pathway.
  • the compound can be, for example, a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4.
  • the TLR4 antagonist can be, for example, an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound that inhibits activity and/or expression of TLR4; an antisense nucleic acid, or an siRNA.
  • Signaling through the TLR:MyD88 pathway can be measured, for example, by assessing the activation status of NFKB or by assessing the level of one or more pro-inflammatory cytokine (e.g., GRO- ⁇ , MCP-I, IL-6).
  • pro-inflammatory cytokine e.g., GRO- ⁇ , MCP-I, IL-6.
  • the compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88.
  • An MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound that inhibits activity and/or expression of MyD88, an antisense nucleic acid, or an siRNA.
  • Also described herein is a method of treating or preventing cancer, such as cancer characterized by elevated expression of MyDSS (e.g., ovarian cancer), in an individual (e.g., a human, such as a woman at risk for or suffering from a cancer characterized by elevated MyD88 expression, such as ovarian cancer), comprising administering to the individual (e.g., a human, such as a woman with ovarian cancer) an effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway.
  • the compound used can be, for example, a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4.
  • the TLR4 antagonist can be, for example, an antibody againstTLR4, a mutated form of TLR4, a mimic of TLR4 a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an antisense nucleic acid, or a SiRNA.
  • the compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88.
  • An MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound that inhibits activity and/or expression of MyD88, an antisense nucleic acid, or an siRNA.
  • a further embodiment of the present invention is a therapeutic compound which inhibits and, preferably, specifically inhibits, signaling through the TLR4:MyD88 pathway, such as a TLR4 antagonist (e.g., a compound that inhibits activity or expression of TLR4).
  • a TLR4 antagonist e.g., a compound that inhibits activity or expression of TLR4
  • the TLR4 antagonist which can be an isolated or substantially pure form, can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an siRNA.
  • the therapeutic compound can be a MyD88 antagonist (e.g., a compound that inhibits activity or expression of MyD88).
  • the MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an siRNA.
  • a therapeutic compound can be used to treat a cancer characterized by elevated MyD88 expression, such as ovarian cancer.
  • a further embodiment of the invention is a method for monitoring drug treatment of an individual who has cancer (e.g., a human, such as a woman with ovarian cancer), comprising: administering a drug to the individual and assaying tissues or cells, such as cancer cells, for MyD88, wherein the expression level of MyD88 is an indicator of the disease status of the individual. For example, a decrease in expression level of MyD88 is typically an indication that the drug treatment is effective.
  • cancer e.g., a human, such as a woman with ovarian cancer
  • the invention relates to a method of enhancing chemo-sensitivity in an individual who has cancer (e.g., a human, such as a woman who has ovarian cancer), in which a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway is administered to an individual in need thereof (such as a woman who has ovarian cancer).
  • cancer e.g., a human, such as a woman who has ovarian cancer
  • a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway is administered to an individual in need thereof (such as a woman who has ovarian cancer).
  • This invention also relates to a method of diagnosing or aiding in diagnosing cancer in an individual (e.g., a human, such as a woman to be diagnosed for ovarian cancer), comprising assaying the expression of MyD88 in a tissue from the individual, wherein the presence of MyD88 or an expression level of MyD88 that is greater than a reference expression level is indicative of tumor tissue or precancerous tissue in the individual.
  • the reference level can be, for example, the level of MyD88 in cells known not to be cancerous, such as cells of the same type (from the same type of organ or tissue) as that being assessed.
  • the level of MyD88 in noncancerous ovarian cells obtained from a woman being assessed or from a different woman or women, can be the reference level.
  • a further embodiment of this invention relates to a method for screening for genetic susceptibility to cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level (an expression level known to be indicative of genetic susceptibility to cancer) of MyD88 that is greater than a reference expression level is indicative of genetic susceptibility to cancer in the individual.
  • the individual being assessed is a human, such as a woman being assessed for genetic susceptibility (likelihood of developing) cancer, such as a genetic susceptibility for developing ovarian cancer.
  • Figure 1 and 2 are Kaplan-Meier curves that illustrate the duration of progression-free interval and overall survival for patients with MyD88+ and MyD88- primary tumors.
  • the pathological diagnosis was papillary serous adenocarcinoma of the ovary for all patients.
  • the clinical course and the response to combination chemotherapy with carboplatin and paclitaxel were markedly different and correlated with MyD88 expression in the analyzed tumors.
  • MyD88 is a cellular marker that can be used to assess the likelihood that an individual will be responsive (will be sensitive or resistant) to chemotherapy with a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel) and whose expression in cancer cells accurately predicts a poor response to such chemotherapy.
  • a plant alkaloid such as a taxane (e.g., paclitaxel or docetaxel) and whose expression in cancer cells accurately predicts a poor response to such chemotherapy.
  • MyD88 is used as a cellular marker to assess the likelihood that ovarian cancer cells will respond (be sensitive or resistant) to treatment with a taxane, such as paclitaxel or docetaxel and to predict a woman's prognosis, in terms of progression-free interval and overall survival.
  • Cancer cells in which MyD 88 is expressed are resistant to taxane chemotherapy, such as treatment with paclitaxel or docetaxel; the prognosis for an individual whose cancer cells express MyD 88 (are MyD 88 " ) is poor (relative to the prognosis of an individual whose cancer cells do not express MyD88 (are MyDSS + ), as assessed, for example, by progression-free interval and overall survival).
  • chemoresistance resistance to a chemotherapeutic drug
  • paclitaxel stimulates cell growth and production of proinflammatory cytokines.
  • the work described herein provides the basis for assessing an individual's responsiveness (sensitivity or resistance) to chemotherapeutic drugs, particularly paclitaxel, as well as for treating or reducing the development of a cancer characterized by elevated expression of MyD88.
  • cancer cells such as ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells to a taxane (paclitaxel, docetaxel).
  • ovarian cancer cells e.g., epithelial ovarian cancer cells
  • breast cancer cells e.g., head and/or neck cancer cells
  • prostate cancer cells non-small cell lung cancer cells
  • small cell lung cancer cells small cell lung cancer cells
  • bladder cancer cells paclitaxel, docetaxel
  • Kaposi's sarcoma cells to a taxane
  • TLR4:MyD88 signaling pathway in cells is a risk factor for developing cancer and a target for reducing (partially or completely) chemoresistance to a taxane, such as resistance to paclitaxel or docetaxel.
  • the cellular marker MyD88 is used to assess the sensitivity/resistance of individuals treated with paclitaxel, a first line chemotherapy agent often used in treating ovarian cancer in women.
  • Applicants have shown that, using the approached described herein, it is possible to determine quickly whether a patient is sensitive to paclitaxel or other taxens.
  • proteins involved in the apoptotic cascade and chemoresistance of cancer cells can be accurately detected. They have exemplified their work using epithelial ovarian cancer cells or EOC (human EOC) isolated from ovarian malignant tumors that were obtained at the time of surgery.
  • the method can be used with a variety of cancer cell types, such as, but not limited to, breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
  • cancer cell types such as, but not limited to, breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
  • tumor specimens are obtained and, optionally, snap- frozen in liquid nitrogen and specimens of appropriate size to provide a sufficient number of cells for analysis (e.g., 8 ⁇ m thick specimens, in the case of ovarian cancer) are stained to facilitate the selection of tumor cells.
  • Target cells are identified based on their immunostaining and collected, such as with the laser capture microdissection (LCM) system described herein. Using CD45 staining proved to be highly useful for the identification and selection of cancer cells from tumor tissue and classification of tumors as either MyD88 positive or negative.
  • the resulting collected cells are lysed and analyzed for MyD88, such as by Western blot or Luminex multiplex assay. In the case of EOC cells, the microdissected cells were evaluated for the expression of MyD88, FasL and XIAP by Western blot analysis.
  • This protocol provides a fast and easy method for analyzing protein expression in tissues, as exemplified by analysis in EOC cells, after LCM. This method was used to evaluate the correlation between MyD88 expression and clinical outcome in women with ovarian cancer. Applicants found that all patients who had MyD88 positive tumors presented with poor progression-free interval and overall survival after chemotherapy with carboplatin and paclitaxel, while the patients with MyD88 negative tumors had an excellent response to chemotherapy. This provides a molecular approach to identify taxane and, specifically, paclitaxel, chemoresistance. Toxicity from agents without therapeutic benefit can be avoided by identifying those patients who will not respond to a specific agent, such as paclitaxel. Molecular markers make it possible to design individualized treatments and improve overall survival. As described herein, Applicants have shown that the status of MyD 88 expression is valuable information to have before beginning chemotherapy with paclitaxel. since that status has an impact on chemo- responsiveness.
  • the present invention relates to a method of assessing responsiveness of a cancer cell to a plant alkaloid, comprising determining whether the cancer cell expresses MyD88 (MyDSe + ) or does not express MyD88 (MyD88 " ), wherein if the cancer cell is MyD88 + , it is resistant to the plant alkaloid and if the cancer cell is MyD88 " , it is sensitive to the plant alkaloid.
  • the plant alkaloid is a taxane, such as paclitaxel or docetaxel. This method is useful to assess the responsiveness of a wide variety of cancer cells (e.g.
  • human cancer cells such as an ovarian cancer cell (such as an EOC cell), a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
  • the invention is also a method of assessing sensitivity or resistance of a cancer cell to chemotherapy with a plant alkaloid, comprising assaying expression of MyD88 in the cancer cell, wherein the cancer cell is sensitive to chemotherapy with the plant alkaloid if the cancer cell does not express MyD88 and the cancer cell is resistant to chemotherapy with the plant alkaloid if the cancer cell expresses MyD88.
  • the plant alkaloid is a taxane, such as paclitaxel or docetaxel.
  • cancer cells e.g., human cells
  • an ovarian cancer cell such as an EOC cell
  • a breast cancer cell such as an EOC cell
  • a head cancer cell such as an EOC cell
  • a neck cancer cell such as an EOC cell
  • a prostate cancer cell such as an EOC cell
  • a non-small cell lung cancer cell such as an EOC cell
  • a small cell lung cancer cell such as an EOC cell
  • a bladder cancer cell such as a stomach cancer cell
  • stomach cancer cell such as a Kaposi's sarcoma cell.
  • Determining whether a cancer cell expresses MyD88 or does not express MyD88 can be carried out by detecting the presence or absence of MyD88 in the cancer cell, detecting the quantity of MyD88 in the cancer cell or detecting nucleic acid that encodes MyD88 in the cancer cell.
  • a further embodiment of the invention is a method of inhibiting proliferation of a cancer cell, comprising contacting the cancer cell with a compound which inhibits signaling through the TLR4:MyD88 pathway.
  • the compound is a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR- 4.
  • the TLR4 antagonist can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA.
  • the compound is a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88.
  • the MyD 88 antagonist can be an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
  • the method can further comprise assessing signaling through the TLR4:MyD88 pathway after the cancer cell is contacted with the compound, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF- ⁇ B or by determining the presence or absence of a pro-inflammatory cytokine or the level of a pro-inflammatory cytokine, such as GRO- ⁇ , MCP-I or IL-6.
  • This method is useful with a variety of cancer (e.g.
  • human cell types such as an ovarian cancer cell (such as an EOC cell), a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
  • an ovarian cancer cell such as an EOC cell
  • a breast cancer cell such as an EOC cell
  • a head cancer cell such as an EOC cell
  • a neck cancer cell such as an EOC cell
  • a prostate cancer cell such as a non-small cell lung cancer cell
  • a small cell lung cancer cell such as a bladder cancer cell
  • stomach cancer cell such as a stomach cancer cell
  • Kaposi's sarcoma cell such as a Kaposi's sarcoma cell.
  • a further embodiment is a method of treating or preventing cancer in an individual, comprising administering to the individual an effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway.
  • the compound can be a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR-4.
  • the TLR4 antagonist can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA.
  • the compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88.
  • the MyD88 antagonist can be selected from an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
  • the method can further comprise assessing signaling through the TLR4:MyD88 pathway after the effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway is administered to the individual, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF- ⁇ B or by determining the presence or absence of a pro-inflammatory cytokine or the level of a pro-inflammatory cytokine, such as GRO- ⁇ , MCP-I or IL- 6.
  • the cancer can be ovarian cancer (epithelial ovarian cancer), breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
  • the cancer typically will be human cancer, but can be cancer in non humans.
  • the cancer is characterized by cancer cells that do not express MyD88 (MyD88 " cells).
  • An isolated therapeutic compound which specifically inhibits signaling through the TLR4:MyD88 pathway is also the subject of this invention. It can be a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4 (e.g., an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound an antisense nucleic acid, and an siRNA).
  • the therapeutic compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88 (e.g., an antibody against MyD88, a mutated form or a mimic of MyD88, a small molecule compound, an antisense nucleic acid, or an siRNA.
  • a method for monitoring drug treatment of an individual who has cancer is also the subject of this invention, the method comprising: administering a drug to the individual; and assaying the expression level of MyD88, wherein the expression level of MyD88 is an indicator of the disease status of the individual.
  • the cancer can be, for example, ovarian cancer, such as human ovarian cancer (e.g., ovarian cancer in which cancer cells do not express MyD88 (MyD88 ⁇ ).
  • the cancer can be breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
  • a subject of this invention is a method of enhancing chemo-sensitivity in an individual who has cancer, comprising administering to an individual in need thereof a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway.
  • the individual might have ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
  • a further subject of the invention is a method of diagnosing or aiding in diagnosing cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD88 which is greater than a reference expression level is indicative of tumor tissue or precancerous tissue in the individual.
  • the subject of this invention is a method for screening for genetic susceptibility to cancer (e.g., ovarian cancer) in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD88 which is greater than a reference expression level is indicative of genetic susceptibility to cancer in the individual.
  • LPS isolated from E. CoIi (0111 :B4), carboplatin and paclitaxel were purchased from Sigma Chemical Co. (St. Louis, MO).
  • the rabbit polyclonal antibody to TLR-4, clone H-80 was purchased from Santa Cruz Biotechnology Inc, (Santa Cruz, CA).
  • Tissue and ascites samples were collected from stage III/IV ovarian cancer patients. Tissues were cut in small aliquots and snap frozen in liquid nitrogen. All patients signed consent forms and the use of patient samples was approved under Yale University's Human Investigations Committee (HIC #10425).
  • EOC cell lines A2780 and CP70 (gifts from Dr. TC Hamilton (16) were propagated in RPMI plus 10% fetal bovine serum (Gemini Bio-Products, Woodland, CA) at 37°C in a 5% CO 2 atmosphere.
  • Primary EOC cells were isolated from malignant ovarian ascites and cultured as previously described (17).
  • EOC cells were isolated from tumors as previously described (17, 18).
  • the normal ovarian surface epithelial cell line immortalized with telomerase was cultured as previously described (19). Purity of the EOC cells was 100% as determined by immuno- staining for cytokeratin antigen.
  • TLR.-4 and MyD88 were evaluated for immunocyto chemistry.
  • the expression and cellular localization of TLR.-4 and MyD88 by EOC cells was performed as previously described (20). In short, sections of tumor samples (5 ⁇ m) were blocked with either 10% horse or goat serum in PBS for 1 hour at room temperature. Following three washes with PBS, samples were incubated overnight at 4°C with either the anti-TLR-4 (Santa Cruz) or the anti- MyD88 antibody. Mouse IgGl or rabbit serum served as negative controls.
  • Cell viability was evaluated using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega, Madison, WI) according to the manufacturer's instructions. The values from the treated cells were compared with the values generated from the untreated cells and reported as percent viability. Each experiment was performed at least three times.
  • cell pellets were processed using the NE-PER Nuclear and Cytoplasmic Extraction Kit (Pierce Biotechnology, Rockford, IL) according to the manufacturer's instructions.
  • the primers used for amplification of human TLR-4 is as follows: (Forward primer) TGGATACGTTTCCTTATAAG ; (Reverse primer)
  • GAAATGGAGGCACCCCTTC 30 cycles of PCR were performed at 95°C for 15 seconds, 54°C for 45 seconds and 72°C for 60 seconds.
  • the size of the product was 449bp.
  • Chemokine production was determined using the Human Cytokine Array kit, III (for cell culture supernatants) (RayBiotech, Atlanta, Georgia) as previously described (20). The intensity of the signals was quantified by densitometry using a digital imaging analysis system and ID Image Analysis Software (Eastman Kodak Company). The signal intensities were normalized against the positive controls on each array membrane, which were given the arbitrary unit of 1. Any expression levels below 0.2 units were considered non-significant.
  • the concentrations of the IL-6, GRO- ⁇ , MCP-I were evaluated by ELISA, according to the manufacturer's instructions. (R&D Systems, Minneapolis, MN.)
  • EOC cells were transiently transfected with a GFP-expressing plasmid containing silencing RNA (siRNA) directed against MyD88 (psiRNA-hMyD88, Invivogen, San Diego, CA). Briefly, 1.5x10 5 cells were seeded in 60mm dishes and cultured overnight until 40-60% confluent. Cells were then transfected for 18h with 2 ⁇ g of DNA using Fugene 6 Transfection reagent (Roche Applied Science, Indianapolis, IN). Ratio of Fugene to DNA was 3:1. Following transfection, cells were allowed to recover in growth media for 24h prior to treatment.
  • siRNA silencing RNA
  • MyD88 psiRNA-hMyD88
  • Transfection was performed using a plasmid containing the full-length cDNA of human MyD88 (pUNO-hMyD88, Invivogen).
  • CP70 and A2780 cells were grown in 75 mm 2 surface tissue culture flasks until they reached 50% confluence.
  • 12 ⁇ l of the Fugene 6 Transfection reagent was added into 4 ml of serum-free media for each transfection.
  • 1 ⁇ g of the plasmid pUNO-hMyD88 was added to the serum-free media containing Fugene 6 Transfection Reagent, and the mixtures were incubated at room temperature for another 15 minutes.
  • the growth media in each flask was discarded, and the corresponding Fugene 6-plasmid-serum-free-media mixture was added and the flasks were incubated overnight at 37°C 5% CO 2 .
  • the transfection media was replaced with fresh growth media the second day and the cells were allowed to recover for 24 hours post transfection before treatments.
  • Eight microns sections were cut with a microtome and fixed on Leica glass foiled PEN- membrane slides. The specimens were fixed in 95% ethanol and H&E stained. Dehydration was performed by immersing the slides in 100% ethanol, followed by Histosolve (Xylene substitute) and air-drying for 10-15 minutes.
  • Leica Laser Capture Microdissection System (Leica Microsystems MA) 6000 ovarian cancer cells were selected and collected in PCR Eppendorf tubes containing Sample Buffer used for preparation of samples for Western blot analysis. Samples underwent five cycles of thawing and freezing followed by 10 minutes at 95°C. Afterward the samples were stored at -2O 0 C until used for Western Blot analysis.
  • TLR-4 expression was evaluated in paraffin sections of ovarian cancer tissues. Positive immunoreactivity for TLR-4 was observed in the tumor cells, but not in the neighboring non-dysplastic cells. The staining was localized in the cytoplasm as well as on the cell surface.
  • TLR-4 expression was evaluated in EOC cell lines isolated from malignant ovarian ascites and from ovarian cancer tissues. Positive immunoreactivity for TLR-4 was observed in all of the evaluated EOC cell lines. The staining pattern was similar to that observed on the paraffin sections. No staining was observed when mouse IgGl was used as negative control.
  • TLR-4 ovarian surface epithelium
  • TLR-4 expressed in EOC cells had the potential to signal and, therefore, be functional
  • MyD88 was cell line and tumor specific.
  • normal OSE cells which expressed TLR- 4 did not express MyD88.
  • TLR-4 and MyD88 were evaluated whether the expression of TLR-4 and MyD88 could be determined in cancer cells isolated with laser microdissector. Ovarian cancer cells were micro-dissected from 8 ⁇ M tissue sections using Laser Capture Micro dissector. Cells were collected in sample buffer and TLR-4 and MyD88 expression was evaluated by Western blot.
  • TLR-4 in EOC samples was established, Applicants evaluated the biological function of this receptor and also evaluated the significance of MyD88 status.
  • EOC cell lines expressing MyD88 (MyD88 + ) and those shown to lack MyD88 (MyD88 " ) were treated with increasing concentrations of LPS (one of the main ligands for TLR-4) for 24h and 48h and cell viability was determined using the CellTiter 96® AQueous One Solution Cell Proliferation Assay. A significant increase in cell proliferation was observed at 24h (p ⁇ 0.01) and 48h (p ⁇ 0.01) in MyD88 + , but not in MyD88 ' EOC cells.
  • LPS induced a time- and dose-dependent increase in cell proliferation in cells expressing MyD88, but not in MyD88- EOC cells. LPS had no effect on viability of OSE cell, which are also negative for MyD88 expression. These results support the dependenceof the proliferative effect of LPS in EOC cells on the presence of MyD88.
  • Example 3 TLR-4 ligation by LPS induced cytokine production in MyD88 expressing cell lines
  • TLR-4 ligation by LPS in cells of the immune system is the induction of cytokine production, mainly of the proinflammatory type.
  • Applicants * next objective was to evaluate whether TLR- 4 ligation by LPS would have a similar effect in EOC cells.
  • MyD88 + EOC cells were incubated in the presence or absence of 10 ⁇ g/ml LPS for 48h and cytokine secretion was evaluated in the cell-free culture supernatants using a human cytokine array.
  • MyD88 EOC cells constitutively secreted a wide range of proinflammatory cytokines and chemokines and this secretion was further enhanced by TLR-4 ligation with LPS.
  • cytokine array led to the identification of specific cytokines/chemokines that were significantly affected by LPS treatment.
  • Applicants performed ELISA and compared the differential response between MyD88 + and MyD88 ' EOC cells. EOC cell lines were treated for 48h with lO ⁇ g/ml LPS and the levels of GRO- ⁇ , MCP-I, and IL-6 were determined. These cytokines were selected because their secretion was shown to be the most significantly affected by LPS treatment. Unstimulated MyD88 + EOC cells constitutively express all the cytokines tested.
  • TLR-4 ligation significantly induced secretion of GRO- ⁇ (p ⁇ 0.05), MCP-I (p ⁇ 0.05) and IL-6 (p ⁇ 0.01).
  • unstimulated MyD88 ' EOC cells secreted undetectable or low levels of these pro-inflammatory cytokines, which were not affected by TLR-4 ligation with LPS.
  • TLR-4 activates the NF- ⁇ B pathway in MyD88 expressing EOC cells
  • NF- ⁇ B is one of the main intracellular pathways mediating the induction of cytokine expression following TLR-4 activation. Therefore Applicants determined if the ligation of TLR-4 induces NF- ⁇ B activation in EOC cells. EOC cells were incubated in the presence or absence of LPS (10 ⁇ g/ml) for I 5 2 and 4h and the activation status of NF- ⁇ B was determined by Western blot analysis. In MyD88 + EOC cells, the p65 active form of NF- ⁇ B was translocated to the nucleus Ih post- treatment with LPS. This shows that in MyD88 + EOC cells, the ligation of TLR-4 by LPS induces NF- ⁇ B activation.
  • Example 5 MyD88 expression was associated with paclitaxel resistance
  • Paclitaxel has been reported to be a potential ligand to TLR-4 (23). Since Applicants' results showed that (i) TLR-4 is ubiquitously expressed in all EOC cell lines tested, (ii) MyD88 expression is cell line specific, and (iii) since paclitaxel resistance is a major characteristic of recurrent ovarian cancer, they evaluated whether MyD88 status affects the response of EOC cells to paclitaxel. EOC cells were treated with 2 ⁇ M paclitaxel for 24h and apoptosis was determined by measuring the activity of caspases -3 and -7 using Caspase-GloTM 3/7 Assay.
  • TLR-4 ligation by paclitaxel would induce the same type of response.
  • MyD88 + and MyD88 " EOC cells were treated with increasing concentrations of paclitaxel for 24h, 48h, and 72h and levels of IL-6 secretion were determined by ELISA.
  • paclitaxel induced a significant increase in IL-6 production in a dose- and time-dependent manner (p ⁇ 0.01).
  • paclitaxel treatment did not affect IL-6 levels in MyD88 " EOC cell lines.
  • Applicants used a high throughput microarray system (Whatman/Scleicher & Schull) to measure the levels of IL-6, IL-4, IL-8, IL-12, RANTES 9 and IFN- ⁇ . Cytokine secretion was measured in the supernatants of EOC cells following 48h incubation with either LPS (lO ⁇ g/ml) or paclitaxel (20 ⁇ M).
  • MyD88 + EOC cells displayed constitutive secretion of the pro-inflammatory cytokines, IL-6, IL-8 and RANTES, and treatment with LPS or paclitaxel resulted in increased secretion of these pro- inflammatory cytokines.
  • MyD88 " EOC cells showed neither constitutive secretion nor LPS- or paclitaxel-induced IL-6, IL-8 or RANTES secretion.
  • Example 7 Expression of MyD88 in EOC cells is necessary for IL-6 production
  • Applicants also determined whether the expression of MyD88 confers resistance that is specific to paclitaxel.
  • Wildtype (wt) A2780 EOC cells (carboplatin sensitive) and A2780 EOC cells transfected with MyD88 were treated with 100 ⁇ g/ml carboplatin or 2 ⁇ M paclitaxel for 48 hours.
  • Apoptosis was determined by measuring caspase-3/7 activity. Wildtype A2780 cells showed significant increase in caspase-3/7 activity following treatment with carboplatin or paclitaxel.
  • MyD88 the cells retained its sensitivity to carboplatin, but not paclitaxel.
  • Example 9 TLR-4 ligation by paclitaxel induces the expression of anti- apoptotic proteins.
  • TLR-4 ligation through NF- ⁇ B and inflammatory molecules promotes cell survival by the induction of the expression of anti-apoptotic proteins (24, 25).
  • XIAP X-linked inhibitor of apoptosis
  • Akt Akt
  • Example 10 Correlation between MyD88 expression in ovarian cancer tissues and patients' progression free survival.
  • Tissue collection was performed prior to chemotherapy and the expression of MyD88 in ovarian cancer tissues was determined by western blot analysis. All patients received six cycles of paclitaxel/carboplatin regimen after surgery.
  • Described herein is a specific defense mechanism used by the innate immune system, which cancer cells have imitated and adapted to generate proinflammatory cytokines, leading to their own proliferation and survival. Described herein is: 1) the expression of TLR-4 in EOC cells; 2) the induction of tumor growth by TLR-4 ligation in MyD88 + EOC cells; 3) the production of chemokines and cytokines by MyD88 + EOC cells upon TLR-4 ligation; and 4) chemo-resistance to paclitaxel mediated by the expression of MyD88.
  • TLRs represent a main receptor pathway, which can induce the expression of pro-inflammatory cytokines (15).
  • TLRs are widely expressed by cells of the immune system and, in response to microbial products or stress factors, initiate an inflammatory process (10).
  • TLRs have been described in non-immune cells, such as mucosal epithelium and trophoblast cells (20, 31). Similar to immune cells, the ligation of TLRs in non-immune cells results in the expression and secretion of pro-inflammatory cytokines (20).
  • Applicants describe the presence of TLR-4 in all EOC cells tested and the differential expression of MyD88.
  • endogenous ligands of TLRs e.g., apoptotic bodies, cellular debris from necrotic cells
  • chemokines can dramatically alter the neoplastic process, not only by recruiting leukocytes that will enhance the inflammatory environment, but also by having a direct effect on nearby stromal and neoplastic cells (32). The induction of these chemokines by TLR-4 activation may, therefore, help to recruit inflammatory cells as well as to enhance tumor growth and neoplastic progression.
  • Melanoma is one example in which chemokines have been shown to exert autocrine control over neoplastic proliferation (33, 34).
  • GRO- ⁇ activity on the CXCR2 receptor attenuates melanoma cell proliferation in vitro, whereas over expression of GRO- ⁇ and GRO- ⁇ enhances tumor colony formation and tumorogenicity in nude mice (34-36).
  • ligation of TLR-4 by LPS induced a significant increase in GRO- ⁇ secretion, which may provide the stimuli for the proliferative effect observed on EOC cells following LPS treatment.
  • pro-inflammatory cytokines and cheniokines has been described as predictors of poor prognosis (37).
  • EOC epithelial ovarian cancer
  • MyD88 is an adaptor protein that is required for Toll-like receptor (TLR) signaling, a signaling pathway involved in inflammatory response to bacterial and viral products.
  • TLR Toll-like receptor
  • MyD88 is a specific marker for paclitaxel resistance and is required for paclitaxel -induced cell growth in EOC cells.
  • the objective of the work described in this example was to develop an optimized method that can detect MyDS 8 expression in ovarian cancer tumors and thus provide a way for MyD88 as a biomarker for selection of therapy.
  • EOC cells were also observed in response to paclitaxel.
  • MyD88 + EOC cells responded to paclitaxel in similar manner as they did with LPS: they produced and secreted pro -inflammatory cytokines. In addition, these cells did not undergo apoptosis in response to paclitaxel. This is in contrast to MyD88 ' cells, which underwent apoptosis and did not secrete cytokine/chemokine in response to paclitaxel.
  • mice by Ding et al showed that similar to LPS, treatment with -paclitaxel activates murine macrophages and induces the secretion of inflammatory cytokines, including TNF ⁇ , IL-6 and IL-8.
  • This effect of paclitaxel was demonstrated to be both TLR-4 and MyD88 dependent (38) (23). These results are supported by the in vivo observation showing a correlation between expression of MyD88 and progression free survival. Although only a small number of patients was evaluated, the data indicate that the expression of MyD88 correlates with a poor survival.
  • TLR-4 ligation is able to induce the activation of the Akt survival pathway and enhance the expression of the anti-apoptotic protein XIAP. Both proteins have been shown to be highly expressed in ovarian cancer and are linked to the develop of chemoresistance (17, 27, 39). Therefore, in MyD88 + EOC cells, the pro-apoptotic effect of paclitaxel is overcome by the induction of the anti-apoptotic proteins (pAKT and XIAP) following TLR-4 ligation. Therefore, in these cells, treatment with paclitaxel does not induce apoptosis but induces the secretion of cytokines/chemokines. The correlation between TLR-4-MyD88 signaling and the upregulation of pAkt and XIAP remains to be determined. NF- ⁇ B may be a link between these pathways.
  • Ovarian cancer tissue samples were collected from advanced stage ovarian cancer patients at the time of surgery. All patients signed consent forms and the use of patient samples was approved under Yale University's Human Investigations Committee (HIC #10425).
  • Tissues were prepared in small aliquots and snap-frozen in liquid N 2 . AU samples were stored in cryovials at -80 0 C until future use.
  • the primary antibody was Dako OV-TL mouse monoclonal cytokeratin-7 from Dakocytomation, Carpinteria, CA.
  • Primary antibody dilution was 1 :150 in 1% BSA mixed in wash buffer.
  • the secondary antibody used was biotinylated anti- mouse IgG (H+L) made in horse from Vector Laboratories, Burlingame, CA. Dilution was 1 :200 in 1% B S A/wash buffer.
  • detection reagent we used Streptavidin-HRP Conjugate from Zymed, San Francisco, CA, in a 1 :300 dilution.
  • the slides were placed inverted into the holder of a Leica AS LMD Laser Microdissection System with the side containing the tissue facing down (non-contact method).
  • Tumor cells were selected according to their morphology for H&E stained samples or according to their immunoreactivity for CK-7 stained samples.
  • lysis of the cells was achieved by submitting the samples to 5 cycles of freezing (liquid N 2 ) and • thawing (95 0 C) followed by boiling for 5 minutes, centrifuged again for 2 minutes and then stored at -8O 0 C until further use.
  • Proteins were resolved under reducing conditions on 12% SDS-PAGE gels and then transferred onto polyvinylidene difluoride paper (NEN Life Sciences, Boston, MA) (5). Membranes were blocked at room temperature for 1 hour with 5% FFPM in PBS/0.05%Tween 20 (PBS-T). After three washes for 10 minutes, each with PBS-T, membranes were incubated overnight at 4 0 C with the primary antibody in PBS-T/1% FFPM. After incubation with the primary antibody, membranes were washed three times with PBS-T and then incubated at room temperature with the appropriate secondary antibody conjugated to peroxidase (Vector Laboratories) in PBS-T/1% FFPM.
  • Results demonstrated that protein expression can be detected by Western blot analysis in microdissected cells obtained from ovarian tumor samples that were snap-frozen in liquid N2. Increasing number of cells were microdissected with a maximum of 8,000 cells and a minimum of 100 cells from 8 ⁇ m ovarian cancer sections. These were analyzed for the expression of MyD88 and other cancer- related proteins, such as XIAP and FasL. A strong signal for MyD88 expression was observed in samples containing 8,000 and 5,000 microdissected cells; no signal was detected for samples containing fewer than 1 ,000 cells.
  • sample thickness affects the signal detected by Western blot was also evaluated. Although increasing the section thickness increased the amount of protein, it affected cutting time and rendered dissection extremely difficult. Therefore, the method was standardized to 8 ⁇ m sections. This allows a quick dissection and provides enough protein to be detected by Western blot.
  • staining specimens e.g. 8 ⁇ m thick specimens to facilitate selection of pure tumor cells
  • target cells e.g., based on their immunostaining and collecting target cells (e.g., with the LCM system)
  • Iy sing collected target cells followed by analysis for protein content/makeup (e.g., by Western blot or Luminex multiplex analysis).
  • Example 12 MyD88 expression is cell-type specific Tumors are heterogeneous tissues containing normal supporting cells, immune cells and cancer cells. Immune cells infiltrate the stroma and may "contaminate" the protein profile of the tumor sample. Indeed, when Applicants analyzed MyD88 expression in H&E stained samples without using appropriate markers for cell type identification, they found that MyD88 was expressed in all samples. The explanation for these results is that the immune cells present in the tumor tissue and surrounding the cancer cells express MyD88. In an H&E stained sample, the selection of specific cells based on their morphology alone is not sufficient to identify intra-tumoral infiltrated cells. An advantage of the LCM is that it affords the possibility of selecting a specific group of cells based on the expression of cellular markers. Therefore, Applicants' next objective was to determine whether immunohistochemistry staining affects the detection of proteins in microdissected samples. Thus, they compared the sensit ) ivity of protein detection of cells obtained
  • Asselin E Mills GB, Tsang BK.
  • XIAP regulates Akt activity and caspase-3- dependent cleavage during cisplatin-induced apoptosis in human ovarian epithelial cancer cells. Cancer Res 2001 ;61 (5): 1862-1868.

Abstract

Described herein is a cellular marker, MyD88, useful for assessing an individual's (patient's) sensitivity (or resistance) to chemotherapy, particularly sensitivity (or resistance) to chemotherapeutic drugs, such as plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel). As described herein, Applicants provide a method by which it is possible to determine whether an individual (cancer cells in an individual) is sensitive to chemotherapy with plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel). Early identification of chemoresistance in patients with cancer is of utmost importance, particularly since it makes it possible to provide the most appropriate therapy.

Description

DRUG RESISTANCE AND METHODS OF REVERSING
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of the filing date of U.S. Provisional Application Serial Number 60/776,304, filed February 23, 2006 and the benefit of the filing date of U.S. Provisional Application Serial Number 60/788,168, filed March 31, 2006. The entire teachings of both referenced applications are incorporated herein by reference.
BACKGROUND OF THE INVENTION
A woman has a 1 in 67 chance of developing ovarian cancer, which is the leading cause of gynecologic cancer deaths. The American Cancer Society estimated that, in 2006, there would be about 20,180 new cases of the disease in the United States and that about 15,310 women would die from the disease that year. Often, ovarian cancer is not diagnosed until it has progressed and the prognosis is not good. The standard first-line chemotherapy for patients with ovarian cancer is a platinum-taxane combination regimen. More than eighty percent of patients will respond initially to this reatement. However, fewer than 10% will remain in remission. Schwartz PE, Cancer Treat Res, 2002; 107:79-118. Improved methods of detecting and treating ovarian cancer are needed.
SUMMARY OF INVENTION
Described herein is a cellular marker, MyD88, useful for assessing an individual's (patient's) sensitivity (or resistance) to chemotherapy, particularly sensitivity (or resistance) to chemotherapeutic drugs, such as plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel). As described herein, Applicants provide a method by which it is possible to determine whether an individual (cancer cells in an individual) is sensitive to chemotherapy with plant alkaloids (e.g., a taxane, such as paclitaxel or docetaxel). Early identification of chemoresi stance in patients with cancer is of utmost importance, particularly since it makes it possible to provide the most appropriate therapy.
Evidence suggests that an inflammatory profile of cytokines and chemokines persisting at a particular site would lead to the development of a chronic disease. Recent studies implicate bacterial infection as one possible link between inflammation and carcinogenesis, but the crucial molecular pathways involved remain unknown. Applicants hypothesized that one upstream signaling pathway that might lead to inflammation in carcinogenesis is mediated by Toll-like receptors (TLRs). Described herein is an adaptive mechanism acquired by ovarian cancer cells that allows them to promote a pro-inflammatory environment and develop chemo-resistance. TLR-4-MyD88 signaling pathway is a risk factor for developing cancer and represents a novel target for the development of bio-modulators.
Provided herein is new evidence that links TLR-4 signaling, inflammation and chemoresistance in ovarian cancer cells. In particular, Applicants demonstrate a link between TLR-4-MyD88 signaling, inflammation, tumor growth and chemoresistance. Described herein is the expression of TLR-4 in cancer cells, as exemplified by epithelial ovarian cancer (EOC) cells and the differential effect of TLR-4 ligation by LPS and paclitaxel in MyD88 positive (MyD88+) and MyD88 negative (MyD88") EOC cells. About 85% to 90% of ovarian cancers are epithelial ovarian carcinomas. The work described herein, such as the link between TLR-4- MyD88 signaling, inflammation, tumor growth and chemoresistance, provides the basis for assessing responsiveness (sensitivity or resistance) to chemotherapeutic drugs, such as a woman's responsiveness (sensitivity or resistance) to chemotherapeutic drugs, particularly paclitaxel, as well as for treating or reducing the development of a cancer characterized by elevated expression of MyD88.
Applicants demonstrate that in EOC cells, signaling through TLR-4 and MyD88 represent a major source of pro-inflammatory cytokines, which promote tumor growth. Furthermore, they have identified a new mechanism for the acquisition of paclitaxel chemo-resistance and therefore, a novel target for the development of molecularly directed therapy in paclitaxel resistant ovarian cancer. In the presence of paclitaxel, EOC cells that express MyD88 show an increase in cell proliferation, secretion and production of proinflammatory cytokines and activation of anti-apoptotic proteins. Cells that do not express MyD88 are paclitaxel sensitive, do not shown NF-κB activation and do not produce inflammatory cytokines.
In one embodiment, this invention relates to a method of assessing the sensitivity or resistance of cancer cells to chemotherapy, particularly chemotherapy with a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel), comprising assaying MyD88 expression in the cancer cells. MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself. Alternatively, nucleic acid encoding MyD88 (e.g., mRNA, DNA) can be assessed. MyD88 expression (a positive result of the analysis) indicates that the individual (patient) from whom the sample was obtained is resistant to such chemotherapy (e.g., chemotherapy with a plant alkaloid, such as a taxane, such as paclitaxel.or docetaxel). Conversely, if the sample is negative for MyD88 expression, the individual (patient) is sensitive to such chemotherapy. For example, if cancer cells (e.g., ovarian cancer cells) obtained from an individual are MyD88 negative (MyD88~), they are sensitive to chemotherapy with a plant alkaloid (e.g., a taxane, such as paclitaxel or docetaxel). Here, the individual's prognosis is good (better than if the patient is resistant to the chemotherapeutic). MyD88 positive cancer cells (e.g., ovarian cancer cells) are resistant to chemotherapy with a plant alkaloid (e.g., a taxane, such as paclitaxel or docetaxel). Alternatively, the level of MyD88 in cancer cells (e.g. in ovarian cancer cells) can be determined and compared with a reference or standard (e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity /resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel). For example, the relationship between MyD88 level in a type of cancer cell (e.g., ovarian cancer cells) and resistance to a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel) can be determined empirically using known methods and a reference or standard established. Resistance/sensitivity to such a chemotherapeutic can be correlated with a range of MyD88 levels and a scale produced. MyD88 level determined in a sample obtained from an individual (e.g., MyD88 level in ovarian cancer cells) can be compared with the correlation or scale and an indication of resistance or sensitivity determined. Alternatively, a "cutoff' MyD88 value (e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel)) can be established and levels determined for cells obtained from a patient compared with that value, to assess their resistance or sensitivity to the plant alkaloid. If the level of MyD88 is greater than the cutoff value, the cancer cells are resistant to such chemotherapy. If the level of MyD 88 is less than the cutoff value, the cells are sensitive to such chemotherapy. The method can be carried out to assess the sensitivity or resistance of an individual suffering from, for example, ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
In a specific embodiment, the invention relates to a method of assessing the sensitivity or resistance of cancer cells to a taxane, such as paclitaxel or docetaxel, used to treat cancer, such as ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma. In this embodiment, the method of assessing the sensitivity or resistance of cancer cells to chemotherapy, particularly chemotherapy with a taxane (e.g., paclitaxel or docetaxel), comprises assaying MyD88 expression in the cancer cells. MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself. Alternatively, nucleic acid encoding MyD88 (e.g., mRNA, DNA) can be assessed. MyD88 expression (a positive result of the analysis) indicates that the individual (patient) from whom the sample was obtained is resistant to such chemotherapy (e.g., chemotherapy with a taxane, such as paclitaxel. or docetaxel). Conversely, if the sample is negative for MyD88 expression, the individual (patient) is sensitive to such chemotherapy. Alternatively, the level of MyD88 in cancer cells (e.g. in ovarian cancer cells) can be determined and compared with a reference or standard (e.g., a correlation between level of MyD88 in cancer cells of the same type as the cells being assessed and the extent/degree of sensitivity/resistance to a chemotherapeutic, such as a correlation between the level of MyD88 in ovarian cancer cells and sensitivity/resistance to paclitaxel). For example, the relationship between MyD88 level in a type of cancer cell (e.g., ovarian cancer cells) and resistance to a taxane (e.g., paclitaxel or docetaxel) can be determined empirically using known methods and a reference or standard established. Resistance/sensitivity to a taxane (e.g., paclitaxel or docetaxel) can be correlated with a range of MyD88 levels and a scale produced. MyD88 level determined in a sample obtained from an individual (e.g., MyD88 level in ovarian cancer cells) can be compared with the correlation or scale and an indication of resistance or sensitivity determined. Alternatively, the reference expression level can be, for example, the level of MyD88 in noncancer cells, such as ovarian cells that are not cancer cells or other cells (e.g., cells from a source other than ovarian tissue), which may or may not be cancer cells. These cells, referred to as reference cells, can be obtained from the individual whose paclitaxel sensitivity /resistance is being assessed or from another individual or individuals. The reference expression levels can be determined at the time an individual is assessed for paclitaxel sensitivity/resistance or can be pre- established reference expression levels (reference expression standard). Alternatively, a "cutoff MyD88 value (e.g., a MyD88 level below which cancer cells are more sensitive than resistant to a taxane (e.g., paclitaxel or docetaxel)) can be established and levels determined for cells obtained from a patient compared with that value, to assess their resistance or sensitivity to the plant alkaloid. If the level of MyD88 is greater than the cutoff value, the cancer cells are resistant to such chemotherapy. If the level of MyD88 is less than the cutoff value, the cells are sensitive to such chemotherapy. The method can be carried out to assess the sensitivity or resistance of ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, or Kaposi's sarcoma cells.
In one embodiment, the invention relates to a method of determining MyD88 (e.g., level of expression, presence or absence of expression) in cells isolated from a tumor, such as by using laser microdissection and dot-blot analysis. In a particular embodiment, laser microdissection is used to obtain a tumor sample that contains a sufficient number of cancer cells to permit assessment of MyD88 (e.g. from about 500 to about 5,000 cells). The sample can be obtained, for example, from a biopsy or a tumor after surgery. Frozen sections can be used, for example. The cells are lysed, using known methods and the product is analyzed (e.g., by Western blot) for MyD88 expression. MyD88 expression can be assessed by, for example, detecting the presence or absence and/or quantity of MyD88 itself. Alternatively, nucleic acid encoding MyD88 (e.g., mRNA, DNA) can be assessed. MyD88 expression (a positive result of the analysis) indicates that the patient from whom the sample was obtained is resistant to paclitaxel. Conversely, if the sample is negative for MyD88 expression, the patient is sensitive to paclitaxel and the patient's prognosis is good (e.g., better than if the patient is resistant to paclitaxel). Samples used for the analysis can be obtained by other methods known to those of skill in the art and can be analyzed for MyD88 expression by other art recognized methods as well.
Also described herein is a method of inhibiting proliferation of a cancer cell, comprising contacting the cell (such as an ovarian cancer cell, particularly human ovarian cancer cells) with a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway. In addition, this invention relates to a method of reducing production of proinflammatory cytokines, which promote tumor growth, by cancer cells and, as a result, reducing the extent to which tumor growth occurs. Such methods can be carried out to inhibit proliferation of cancer cells of many types in which there is an active TLR4:MyD88 signaling pathway, including, but not limited to, ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
This invention is also a method of reducing (partially or totally) tumor growth, comprising contacting tumor cells in which there is an active TLR4:MyD88 signaling pathway with a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 signaling pathway. Also the subject of this invention is a method of treating or preventing cancer, such as a cancer characterized by an active TLR4:MyD88 signaling pathway, in an individual in need thereof, comprising administering to the individual (e.g., a human suffering from or at risk of developing such a cancer) an effective amount of a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway. In a specific embodiment, the invention is a method of treating or preventing ovarian cancer, in an individual (such as a woman suffering from or at risk of developing ovarian cancer characterized by an active TLR4:MyD88 signaling pathway), comprising administering to the individual (e.g., the woman) an effective amount of a compound that inhibits, directly or indirectly, signaling through the TLR4:MyD88 pathway. Also described herein is a method for inhibiting proliferation of a cancer cell, comprising contacting the cell (such as an ovarian cancer cell, particularly human ovarian cancer cells) with a compound that inhibits signaling through the TLR4:MyD88 pathway. The compound can be, for example, a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4. The TLR4 antagonist can be, for example, an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound that inhibits activity and/or expression of TLR4; an antisense nucleic acid, or an siRNA. Signaling through the TLR:MyD88 pathway can be measured, for example, by assessing the activation status of NFKB or by assessing the level of one or more pro-inflammatory cytokine (e.g., GRO-α, MCP-I, IL-6). Alternatively, the compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88. An MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound that inhibits activity and/or expression of MyD88, an antisense nucleic acid, or an siRNA.
Also described herein is a method of treating or preventing cancer, such as cancer characterized by elevated expression of MyDSS (e.g., ovarian cancer), in an individual (e.g., a human, such as a woman at risk for or suffering from a cancer characterized by elevated MyD88 expression, such as ovarian cancer), comprising administering to the individual (e.g., a human, such as a woman with ovarian cancer) an effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway. In the method, the compound used can be, for example, a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4. The TLR4 antagonist can be, for example, an antibody againstTLR4, a mutated form of TLR4, a mimic of TLR4 a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an antisense nucleic acid, or a SiRNA. Alternatively, the compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88. An MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound that inhibits activity and/or expression of MyD88, an antisense nucleic acid, or an siRNA. A further embodiment of the present invention is a therapeutic compound which inhibits and, preferably, specifically inhibits, signaling through the TLR4:MyD88 pathway, such as a TLR4 antagonist (e.g., a compound that inhibits activity or expression of TLR4). The TLR4 antagonist, which can be an isolated or substantially pure form, can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an siRNA. Alternatively, the therapeutic compound can be a MyD88 antagonist (e.g., a compound that inhibits activity or expression of MyD88). The MyD88 antagonist can be, for example, an antibody against MyD88, a mutated form of MyD88, a small molecule compound that inhibits activity and/or expression of TLR4, an antisense nucleic acid, or an siRNA. A therapeutic compound can be used to treat a cancer characterized by elevated MyD88 expression, such as ovarian cancer.
A further embodiment of the invention is a method for monitoring drug treatment of an individual who has cancer (e.g., a human, such as a woman with ovarian cancer), comprising: administering a drug to the individual and assaying tissues or cells, such as cancer cells, for MyD88, wherein the expression level of MyD88 is an indicator of the disease status of the individual. For example, a decrease in expression level of MyD88 is typically an indication that the drug treatment is effective.
In another embodiment, the invention relates to a method of enhancing chemo-sensitivity in an individual who has cancer (e.g., a human, such as a woman who has ovarian cancer), in which a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway is administered to an individual in need thereof (such as a woman who has ovarian cancer).
This invention also relates to a method of diagnosing or aiding in diagnosing cancer in an individual (e.g., a human, such as a woman to be diagnosed for ovarian cancer), comprising assaying the expression of MyD88 in a tissue from the individual, wherein the presence of MyD88 or an expression level of MyD88 that is greater than a reference expression level is indicative of tumor tissue or precancerous tissue in the individual. The reference level can be, for example, the level of MyD88 in cells known not to be cancerous, such as cells of the same type (from the same type of organ or tissue) as that being assessed. For example, the level of MyD88 in noncancerous ovarian cells, obtained from a woman being assessed or from a different woman or women, can be the reference level.
A further embodiment of this invention relates to a method for screening for genetic susceptibility to cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level (an expression level known to be indicative of genetic susceptibility to cancer) of MyD88 that is greater than a reference expression level is indicative of genetic susceptibility to cancer in the individual. In a specific embodiment, the individual being assessed is a human, such as a woman being assessed for genetic susceptibility (likelihood of developing) cancer, such as a genetic susceptibility for developing ovarian cancer.
BRIEF DESCRIPTION OF THE DRAWINGS
The following figures (Figure 1 and 2) are Kaplan-Meier curves that illustrate the duration of progression-free interval and overall survival for patients with MyD88+ and MyD88- primary tumors. The pathological diagnosis was papillary serous adenocarcinoma of the ovary for all patients. However, the clinical course and the response to combination chemotherapy with carboplatin and paclitaxel were markedly different and correlated with MyD88 expression in the analyzed tumors.
Figure 1 : Mean progression-free interval was zero for patients with MyD88+ tumors (n = 7) and 35 months for patients with MyD88- tumors (n = 5) treated with carboplatin and paclitaxel. This difference was statistically significant (log rank statistical test p = 0.009).
Figure 2: Overall survival according to the MyD88 status of the analyzed tumors shows divergent Kaplan-Meier curves and was statistically significant (-2 log likelihood ratio test p = 0.022). DETAILED DESCRIPTION OF THE INVENTION
As described herein, MyD88 is a cellular marker that can be used to assess the likelihood that an individual will be responsive (will be sensitive or resistant) to chemotherapy with a plant alkaloid, such as a taxane (e.g., paclitaxel or docetaxel) and whose expression in cancer cells accurately predicts a poor response to such chemotherapy. In a specific embodiment, MyD88 is used as a cellular marker to assess the likelihood that ovarian cancer cells will respond (be sensitive or resistant) to treatment with a taxane, such as paclitaxel or docetaxel and to predict a woman's prognosis, in terms of progression-free interval and overall survival. Cancer cells in which MyD 88 is expressed are resistant to taxane chemotherapy, such as treatment with paclitaxel or docetaxel; the prognosis for an individual whose cancer cells express MyD 88 (are MyD 88" ) is poor (relative to the prognosis of an individual whose cancer cells do not express MyD88 (are MyDSS+), as assessed, for example, by progression-free interval and overall survival). As also described herein, Applicants have shown that an adaptive mechanism present in cancer cells allows the cells to exhibit chemoresistance (resistance to a chemotherapeutic drug), particularly resistance to paclitaxel and more particularly resistance of ovarian cancer cells to paclitaxel. They have also shown that, in MyD88 positive cancer cells, such as in ovarian cancer cells, paclitaxel stimulates cell growth and production of proinflammatory cytokines. The work described herein, such as the link between TLR-4-MyD88 signaling, inflammation, tumor growth and chemoresistance, provides the basis for assessing an individual's responsiveness (sensitivity or resistance) to chemotherapeutic drugs, particularly paclitaxel, as well as for treating or reducing the development of a cancer characterized by elevated expression of MyD88. It provides the basis for assessing the responsiveness of cancer cells, such as ovarian cancer cells (e.g., epithelial ovarian cancer cells), breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells to a taxane (paclitaxel, docetaxel). It is reasonable to conclude that the TLR4:MyD88 signaling pathway in cells is a risk factor for developing cancer and a target for reducing (partially or completely) chemoresistance to a taxane, such as resistance to paclitaxel or docetaxel. In one embodiment, the cellular marker MyD88 is used to assess the sensitivity/resistance of individuals treated with paclitaxel, a first line chemotherapy agent often used in treating ovarian cancer in women. Applicants have shown that, using the approached described herein, it is possible to determine quickly whether a patient is sensitive to paclitaxel or other taxens. Applicants have demonstrated that proteins involved in the apoptotic cascade and chemoresistance of cancer cells can be accurately detected. They have exemplified their work using epithelial ovarian cancer cells or EOC (human EOC) isolated from ovarian malignant tumors that were obtained at the time of surgery. The method can be used with a variety of cancer cell types, such as, but not limited to, breast cancer cells, head and/or neck cancer cells, prostate cancer cells, non-small cell lung cancer cells, small cell lung cancer cells, bladder cancer cells, stomach cancer cells, and Kaposi's sarcoma cells.
In one embodiment, tumor specimens are obtained and, optionally, snap- frozen in liquid nitrogen and specimens of appropriate size to provide a sufficient number of cells for analysis (e.g., 8 μm thick specimens, in the case of ovarian cancer) are stained to facilitate the selection of tumor cells. Target cells are identified based on their immunostaining and collected, such as with the laser capture microdissection (LCM) system described herein. Using CD45 staining proved to be highly useful for the identification and selection of cancer cells from tumor tissue and classification of tumors as either MyD88 positive or negative. The resulting collected cells are lysed and analyzed for MyD88, such as by Western blot or Luminex multiplex assay. In the case of EOC cells, the microdissected cells were evaluated for the expression of MyD88, FasL and XIAP by Western blot analysis.
This protocol provides a fast and easy method for analyzing protein expression in tissues, as exemplified by analysis in EOC cells, after LCM. This method was used to evaluate the correlation between MyD88 expression and clinical outcome in women with ovarian cancer. Applicants found that all patients who had MyD88 positive tumors presented with poor progression-free interval and overall survival after chemotherapy with carboplatin and paclitaxel, while the patients with MyD88 negative tumors had an excellent response to chemotherapy. This provides a molecular approach to identify taxane and, specifically, paclitaxel, chemoresistance. Toxicity from agents without therapeutic benefit can be avoided by identifying those patients who will not respond to a specific agent, such as paclitaxel. Molecular markers make it possible to design individualized treatments and improve overall survival. As described herein, Applicants have shown that the status of MyD 88 expression is valuable information to have before beginning chemotherapy with paclitaxel. since that status has an impact on chemo- responsiveness.
The present invention relates to a method of assessing responsiveness of a cancer cell to a plant alkaloid, comprising determining whether the cancer cell expresses MyD88 (MyDSe+) or does not express MyD88 (MyD88"), wherein if the cancer cell is MyD88+, it is resistant to the plant alkaloid and if the cancer cell is MyD88", it is sensitive to the plant alkaloid. In one embodiment, the plant alkaloid is a taxane, such as paclitaxel or docetaxel. This method is useful to assess the responsiveness of a wide variety of cancer cells (e.g. human cancer cells) such as an ovarian cancer cell (such as an EOC cell), a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
The invention is also a method of assessing sensitivity or resistance of a cancer cell to chemotherapy with a plant alkaloid, comprising assaying expression of MyD88 in the cancer cell, wherein the cancer cell is sensitive to chemotherapy with the plant alkaloid if the cancer cell does not express MyD88 and the cancer cell is resistant to chemotherapy with the plant alkaloid if the cancer cell expresses MyD88. In one embodiment, the plant alkaloid is a taxane, such as paclitaxel or docetaxel. This methodis useful with a wide variety of cancer cells (e.g., human cells), such as an ovarian cancer cell (such as an EOC cell), a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
Determining whether a cancer cell expresses MyD88 or does not express MyD88 can be carried out by detecting the presence or absence of MyD88 in the cancer cell, detecting the quantity of MyD88 in the cancer cell or detecting nucleic acid that encodes MyD88 in the cancer cell. A further embodiment of the invention is a method of inhibiting proliferation of a cancer cell, comprising contacting the cancer cell with a compound which inhibits signaling through the TLR4:MyD88 pathway. In one embodiment, the compound is a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR- 4. The TLR4 antagonist can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA. In a further embodiment, the compound is a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88. The MyD 88 antagonist can be an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA. The method can further comprise assessing signaling through the TLR4:MyD88 pathway after the cancer cell is contacted with the compound, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF-κB or by determining the presence or absence of a pro-inflammatory cytokine or the level of a pro-inflammatory cytokine, such as GRO-α, MCP-I or IL-6. This method is useful with a variety of cancer (e.g. human) cell types, such as an ovarian cancer cell (such as an EOC cell), a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
A further embodiment is a method of treating or preventing cancer in an individual, comprising administering to the individual an effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway. The compound can be a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR-4. The TLR4 antagonist can be an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA. The compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88. The MyD88 antagonist can be selected from an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
The method can further comprise assessing signaling through the TLR4:MyD88 pathway after the effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway is administered to the individual, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF-κB or by determining the presence or absence of a pro-inflammatory cytokine or the level of a pro-inflammatory cytokine, such as GRO-α, MCP-I or IL- 6. In this embodiment, the cancer can be ovarian cancer (epithelial ovarian cancer), breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma. The cancer typically will be human cancer, but can be cancer in non humans. In specific embodiments, the cancer is characterized by cancer cells that do not express MyD88 (MyD88" cells).
An isolated therapeutic compound which specifically inhibits signaling through the TLR4:MyD88 pathway is also the subject of this invention. It can be a TLR4 antagonist, such as a compound that inhibits activity or expression of TLR4 (e.g., an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound an antisense nucleic acid, and an siRNA). The therapeutic compound can be a MyD88 antagonist, such as a compound that inhibits activity or expression of MyD88 (e.g., an antibody against MyD88, a mutated form or a mimic of MyD88, a small molecule compound, an antisense nucleic acid, or an siRNA.
A method for monitoring drug treatment of an individual who has cancer is also the subject of this invention, the method comprising: administering a drug to the individual; and assaying the expression level of MyD88, wherein the expression level of MyD88 is an indicator of the disease status of the individual. The cancer can be, for example, ovarian cancer, such as human ovarian cancer (e.g., ovarian cancer in which cancer cells do not express MyD88 (MyD88~). Alternatively the cancer can be breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma. Also a subject of this invention is a method of enhancing chemo-sensitivity in an individual who has cancer, comprising administering to an individual in need thereof a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway. The individual might have ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
A further subject of the invention is a method of diagnosing or aiding in diagnosing cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD88 which is greater than a reference expression level is indicative of tumor tissue or precancerous tissue in the individual. Also the subject of this invention is a method for screening for genetic susceptibility to cancer (e.g., ovarian cancer) in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD88 which is greater than a reference expression level is indicative of genetic susceptibility to cancer in the individual.
The present invention is illustrated by the following examples, which are not intended to be limiting in any way.
MATERIALS AND METHODS
The following materials and methods were used in the work described in Examples 1-10.
Reagents
LPS isolated from E. CoIi (0111 :B4), carboplatin and paclitaxel were purchased from Sigma Chemical Co. (St. Louis, MO). The rabbit polyclonal antibody to TLR-4, clone H-80 was purchased from Santa Cruz Biotechnology Inc, (Santa Cruz, CA).
Patients & samples
Tissue and ascites samples were collected from stage III/IV ovarian cancer patients. Tissues were cut in small aliquots and snap frozen in liquid nitrogen. All patients signed consent forms and the use of patient samples was approved under Yale University's Human Investigations Committee (HIC #10425).
Cell lines
Human EOC cell lines, A2780 and CP70 (gifts from Dr. TC Hamilton (16) were propagated in RPMI plus 10% fetal bovine serum (Gemini Bio-Products, Woodland, CA) at 37°C in a 5% CO2 atmosphere. Primary EOC cells were isolated from malignant ovarian ascites and cultured as previously described (17). EOC cells were isolated from tumors as previously described (17, 18). The normal ovarian surface epithelial cell line immortalized with telomerase was cultured as previously described (19). Purity of the EOC cells was 100% as determined by immuno- staining for cytokeratin antigen.
Imiminohistochemistry
Twenty four samples of ovarian cancer tissues were evaluated for immunocyto chemistry. The expression and cellular localization of TLR.-4 and MyD88 by EOC cells was performed as previously described (20). In short, sections of tumor samples (5μm) were blocked with either 10% horse or goat serum in PBS for 1 hour at room temperature. Following three washes with PBS, samples were incubated overnight at 4°C with either the anti-TLR-4 (Santa Cruz) or the anti- MyD88 antibody. Mouse IgGl or rabbit serum served as negative controls. After three washes with PBS, specific staining was detected by incubating with either a peroxidase-conjugated horse anti-mouse antibody (1:1000 dilution) or a peroxidase- conjugated goat anti-rabbit antibody (1 : 1000 dilution) for 1 hour followed by a five- minute incubation with DAB substrate (Vector Laboratories). Cells and tissue sections were then counterstained with haematoxylin (Sigma Chemical Co.) before dehydration with ethanol and Histosolve (Shandon Inc., Pittsburg, PA). Slides were then mounted with Permount (Fisher Scientific, Pittsburg, PA) and visualized by light microscopy.
Cell viability assay
Cell viability was evaluated using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega, Madison, WI) according to the manufacturer's instructions. The values from the treated cells were compared with the values generated from the untreated cells and reported as percent viability. Each experiment was performed at least three times.
Protein preparation
Protein was extracted as previously described (17). Briefly, cell pellets were lysed in a cold solution of 1% NP40 and 0.1% SDS in PBS, with freshly added Protease Inhibitor Cocktail (20 μl/ml; Sigma Chemical Co.) and 2 mM PMSF (Sigma Chemical Co.). Protein concentration was determined by BioRad Protein Assay (BioRad, Hercules, CA) and proteins were stored at -4O0C until further use.
For separation of the cytoplasmic and nuclear fractions, cell pellets were processed using the NE-PER Nuclear and Cytoplasmic Extraction Kit (Pierce Biotechnology, Rockford, IL) according to the manufacturer's instructions.
SDS-PAGE and Western blots
Twenty μg protein were denatured in sample buffer (2.5% SDS, 10% glycerol, 5% β-mercapto-ethanol, 0.15 M Tris (pH=6.8) and 0.01% bromophenol blue) and subjected to 12% SDS-PAGE as previously described (17). Antibodies used: rabbit anti-TLR-4 (Santa Cruz Biotechnology, Santa Cruz, CA, 1: 1,000), rabbit anti-MyD88 (eBiosciences, San Diego, CA, 1 :1,000), mouse anti-NF-κB (Santa Cruz Biotechnology, 1 : 1,000), mouse anti-DNA topoisomerase 1 (BD Biosciences, San Jose, CA, 1:500) and, rabbit anti-actin (Sigma Chemical Co., 1 : 10,000). Proteins were visualized using enhanced chemiluminescence (Pierce Biotechnology).
RNA isolation and RT-PCR
Total RNA was isolated using the RNeasy Mini Kit (Qiagen, Valencia, CA) according to the manufacturer's instructions. Reverse transcription was performed on 5 μg of total RNA using the First Strand cDNA Synthesis kit (Amersham Biosciences, Buckinghamshire, U.K.) according to the manufacturer's instructions. The primers used for amplification of human TLR-4 is as follows: (Forward primer) TGGATACGTTTCCTTATAAG ; (Reverse primer)
GAAATGGAGGCACCCCTTC. 30 cycles of PCR were performed at 95°C for 15 seconds, 54°C for 45 seconds and 72°C for 60 seconds. The size of the product was 449bp.
Caspase-GIo™ 3/7 assay
Ten μg of protein in a 50 μl total volume was mixed with 50 μl of equilibrated Caspase-GIo™ 3/7 reagents (Promega). After incubating at room temperature for Ih, luminescence was measured using TD 20/20 Luminometer (Turner Designs, Sunnyvale, CA). Blank values were subtracted and fold-increase in activity was calculated based on activity measured from untreated cells. Each sample was measured in triplicate.
Cytokine and Chemokine studies
Chemokine production was determined using the Human Cytokine Array kit, III (for cell culture supernatants) (RayBiotech, Atlanta, Georgia) as previously described (20). The intensity of the signals was quantified by densitometry using a digital imaging analysis system and ID Image Analysis Software (Eastman Kodak Company). The signal intensities were normalized against the positive controls on each array membrane, which were given the arbitrary unit of 1. Any expression levels below 0.2 units were considered non-significant.
The concentrations of the IL-6, GRO-α, MCP-I were evaluated by ELISA, according to the manufacturer's instructions. (R&D Systems, Minneapolis, MN.)
A panel of 10 cytokines, pro and anti-inflammatory, was simultaneously evaluated using the Fast Quant Human II kit (Whatman/Schleicher & Schuell) according to the manufacturer instructions. The signal was detected using the Genepix 4000 microarray reader and the results were analyzed using the Genepix 3.0 software (Whatman/Schleicher & Schuell).
RNA interference
EOC cells were transiently transfected with a GFP-expressing plasmid containing silencing RNA (siRNA) directed against MyD88 (psiRNA-hMyD88, Invivogen, San Diego, CA). Briefly, 1.5x105 cells were seeded in 60mm dishes and cultured overnight until 40-60% confluent. Cells were then transfected for 18h with 2μg of DNA using Fugene 6 Transfection reagent (Roche Applied Science, Indianapolis, IN). Ratio of Fugene to DNA was 3:1. Following transfection, cells were allowed to recover in growth media for 24h prior to treatment.
MyD88 Transfections
Transfection was performed using a plasmid containing the full-length cDNA of human MyD88 (pUNO-hMyD88, Invivogen). CP70 and A2780 cells were grown in 75 mm2 surface tissue culture flasks until they reached 50% confluence. 12 μl of the Fugene 6 Transfection reagent was added into 4 ml of serum-free media for each transfection. After 5 minutes of incubation at room temperature, 1 μg of the plasmid pUNO-hMyD88 was added to the serum-free media containing Fugene 6 Transfection Reagent, and the mixtures were incubated at room temperature for another 15 minutes. The growth media in each flask was discarded, and the corresponding Fugene 6-plasmid-serum-free-media mixture was added and the flasks were incubated overnight at 37°C 5% CO2. The transfection media was replaced with fresh growth media the second day and the cells were allowed to recover for 24 hours post transfection before treatments.
Laser Capture Microdissection (LCM)
Ovarian cancer specimens (n=24) obtained in the operating room were snap- frozen in liquid nitrogen and stored in cryovials at -800C. For Laser capture microdissection, a small fraction of tissue was embedded in OCT at -200C. Eight microns sections were cut with a microtome and fixed on Leica glass foiled PEN- membrane slides. The specimens were fixed in 95% ethanol and H&E stained. Dehydration was performed by immersing the slides in 100% ethanol, followed by Histosolve (Xylene substitute) and air-drying for 10-15 minutes.
Using the Leica Laser Capture Microdissection System (Leica Microsystems MA) 6000 ovarian cancer cells were selected and collected in PCR Eppendorf tubes containing Sample Buffer used for preparation of samples for Western blot analysis. Samples underwent five cycles of thawing and freezing followed by 10 minutes at 95°C. Afterward the samples were stored at -2O0C until used for Western Blot analysis.
Statistical analysis
Data are expressed as mean ± standard deviation (21). Statistical significance (p < 0.05) was determined using one-way ANOVA with the Bonferonni correction. Survival curve of the patients were done by the method of Kaplan-Meier (22) and the significance of the difference was estimated by log — rank test. Example 1: EOC expression of TLR-4 and the signaling adapter protein MyD88
First, Applicants determined whether TLR-4 is expressed in EOC cells. TLR-4 expression was evaluated in paraffin sections of ovarian cancer tissues. Positive immunoreactivity for TLR-4 was observed in the tumor cells, but not in the neighboring non-dysplastic cells. The staining was localized in the cytoplasm as well as on the cell surface. Next, TLR-4 expression was evaluated in EOC cell lines isolated from malignant ovarian ascites and from ovarian cancer tissues. Positive immunoreactivity for TLR-4 was observed in all of the evaluated EOC cell lines. The staining pattern was similar to that observed on the paraffin sections. No staining was observed when mouse IgGl was used as negative control.
To confirm the specificity of these findings, the expression of TLR-4 by EOC cells and tumors was further evaluated by RT-PCR and Western blot analysis. EOC cells and tumors expressed TLR-4, both at the mRNA and protein level. In addition, the mRNA message for TLR-4 was found to be expressed in a normal ovarian surface epithelium (OSE) cell line immortalized with telomerase (19).
In order to determine whether TLR-4 expressed in EOC cells had the potential to signal and, therefore, be functional, Applicants evaluated expression of the TLR signaling adapter protein, MyD88. Unlike TLR-4, which was ubiquitously expressed by all EOC cell lines and tumors evaluated, the expression of MyD88 was cell line and tumor specific. Furthermore, normal OSE cells, which expressed TLR- 4 did not express MyD88.
Applicants also evaluated whether the expression of TLR-4 and MyD88 could be determined in cancer cells isolated with laser microdissector. Ovarian cancer cells were micro-dissected from 8 μM tissue sections using Laser Capture Micro dissector. Cells were collected in sample buffer and TLR-4 and MyD88 expression was evaluated by Western blot.
Example 2: MyD88 expression is required for LPS-induced tumor growth
Once the expression of TLR-4 in EOC samples was established, Applicants evaluated the biological function of this receptor and also evaluated the significance of MyD88 status. EOC cell lines expressing MyD88 (MyD88+) and those shown to lack MyD88 (MyD88") were treated with increasing concentrations of LPS (one of the main ligands for TLR-4) for 24h and 48h and cell viability was determined using the CellTiter 96® AQueous One Solution Cell Proliferation Assay. A significant increase in cell proliferation was observed at 24h (p<0.01) and 48h (p<0.01) in MyD88+, but not in MyD88' EOC cells. Thus, LPS induced a time- and dose- dependent increase in cell proliferation in cells expressing MyD88, but not in MyD88- EOC cells. LPS had no effect on viability of OSE cell, which are also negative for MyD88 expression. These results support the dependenceof the proliferative effect of LPS in EOC cells on the presence of MyD88.
Example 3: TLR-4 ligation by LPS induced cytokine production in MyD88 expressing cell lines
One of the main characteristics of TLR-4 ligation by LPS in cells of the immune system is the induction of cytokine production, mainly of the proinflammatory type. Thus, Applicants* next objective was to evaluate whether TLR- 4 ligation by LPS would have a similar effect in EOC cells. Thus, MyD88+ EOC cells were incubated in the presence or absence of 10 μg/ml LPS for 48h and cytokine secretion was evaluated in the cell-free culture supernatants using a human cytokine array. MyD88 EOC cells constitutively secreted a wide range of proinflammatory cytokines and chemokines and this secretion was further enhanced by TLR-4 ligation with LPS.
The results from the cytokine array led to the identification of specific cytokines/chemokines that were significantly affected by LPS treatment. To validate these results, Applicants performed ELISA and compared the differential response between MyD88+ and MyD88' EOC cells. EOC cell lines were treated for 48h with lOμg/ml LPS and the levels of GRO-α, MCP-I, and IL-6 were determined. These cytokines were selected because their secretion was shown to be the most significantly affected by LPS treatment. Unstimulated MyD88+ EOC cells constitutively express all the cytokines tested. In addition, TLR-4 ligation significantly induced secretion of GRO-α (p< 0.05), MCP-I (p<0.05) and IL-6 (p<0.01). In contrast, unstimulated MyD88' EOC cells secreted undetectable or low levels of these pro-inflammatory cytokines, which were not affected by TLR-4 ligation with LPS. These findings suggest that the increased production of proinflammatory cytokines in response to LPS was dependent on MyD88 expression in EOC cells.
Example 4: TLR-4 activates the NF-κB pathway in MyD88 expressing EOC cells
NF-κB is one of the main intracellular pathways mediating the induction of cytokine expression following TLR-4 activation. Therefore Applicants determined if the ligation of TLR-4 induces NF-κB activation in EOC cells. EOC cells were incubated in the presence or absence of LPS (10 μg/ml) for I5 2 and 4h and the activation status of NF-κB was determined by Western blot analysis. In MyD88+ EOC cells, the p65 active form of NF-κB was translocated to the nucleus Ih post- treatment with LPS. This shows that in MyD88+ EOC cells, the ligation of TLR-4 by LPS induces NF-κB activation. Interestingly, constitutive nuclear localization of p65 NF-κB was observed in MyD88" EOC cells, and its level was not affected by LPS treatment. Therefore, Applicants determined the status of the NF-κ inhibitor, IκB-α, in these cells. Results show IκB-α degradation in MyD88+ cells in response to LPS. In contrast, constitutive degradation of IκB-oc was not observed in MyD88" cells and in addition, no change in its level was observed after LPS treatment. These results are evidence that TLR-4 ligation by LPS results in early phase activation of NF-κB in MyD88+ but not in MyD88" EOC cells.
Example 5: MyD88 expression was associated with paclitaxel resistance
Paclitaxel has been reported to be a potential ligand to TLR-4 (23). Since Applicants' results showed that (i) TLR-4 is ubiquitously expressed in all EOC cell lines tested, (ii) MyD88 expression is cell line specific, and (iii) since paclitaxel resistance is a major characteristic of recurrent ovarian cancer, they evaluated whether MyD88 status affects the response of EOC cells to paclitaxel. EOC cells were treated with 2 μM paclitaxel for 24h and apoptosis was determined by measuring the activity of caspases -3 and -7 using Caspase-Glo™ 3/7 Assay. There was a significant increase in caspase- 3/7 activity following paclitaxel treatment in MyD88" EOC cell lines. Significant increase in caspase activity was also observed in normal OSE cells following paclitaxel treatment. In contrast, no significant change in caspase- 3/7 activity was detected in MyD88+ EOC cells. These results are evidence that the MyD88 status of EOC cells determines their apoptotic response to paclitaxel. Example 6: MyD88 expression mediates Paclitaxel-induced production of proinflammatory cytokines.
Since the previous results showed that TLR-4 ligation by LPS in MyD88+ EOC cells results in increased cytokine secretion, Applicants then determined if TLR-4 ligation by paclitaxel would induce the same type of response. MyD88+ and MyD88" EOC cells were treated with increasing concentrations of paclitaxel for 24h, 48h, and 72h and levels of IL-6 secretion were determined by ELISA. In MyD88+ EOC cell lines, paclitaxel induced a significant increase in IL-6 production in a dose- and time-dependent manner (p<0.01). In contrast, paclitaxel treatment did not affect IL-6 levels in MyD88" EOC cell lines.
To further characterize the effect of paclitaxel and LPS on the secretion of several cytokines and chemokines in MyD88+ and MyD88" EOC cells, Applicants used a high throughput microarray system (Whatman/Scleicher & Schull) to measure the levels of IL-6, IL-4, IL-8, IL-12, RANTES9 and IFN- γ. Cytokine secretion was measured in the supernatants of EOC cells following 48h incubation with either LPS (lOμg/ml) or paclitaxel (20μM). MyD88+ EOC cells displayed constitutive secretion of the pro-inflammatory cytokines, IL-6, IL-8 and RANTES, and treatment with LPS or paclitaxel resulted in increased secretion of these pro- inflammatory cytokines. In contrast, MyD88" EOC cells showed neither constitutive secretion nor LPS- or paclitaxel-induced IL-6, IL-8 or RANTES secretion. Interestingly, neither MyD88+ nor MyD88" EOC cells secreted detectable levels of IL-4, IL-12 and IFN-γ, with or without treatment with either LPS or paclitaxel. Example 7: Expression of MyD88 in EOC cells is necessary for IL-6 production
In order to confirm that in MyD88+ EOC cells, the ligation of TLR-4 by either LPS or paclitaxel induces the production of cytokines, Applicants inhibited its expression in MyD88+ Rl 82 EOC cells using RNA interference technology. As shown above, wild type (wt) Rl 82 cells produce high levels of IL-6 in response to both LPS (lOμg/ml) and paclitaxel (2μM). However, following the inhibition of MyD88 expression by RNAi, treatment with LPS or paclitaxel resulted in decreased IL-6 secretion. These findings confirmed that the cytokine response in EOC cells following LPS and paclitaxel treatment involves signaling through MyD88. Example 8: Induction of MyD88 expression in EOC cells reverses chemo- sensitivity to paclitaxel.
Because Appliants observed a positive correlation between MyD88 expression and resistance to paclitaxel-induced apoptosis, their next objective was to determine whether the induction of MyD88 expression in EOC might confer resistance to paclitaxel. Therefore, they induced the expression of MyD88 in A2780 and CP70 EOC cells, both MyD88" and paclitaxel-sensitive, by transient transfection with a plasmid vector containing the full-length cDNA of MyD 88 (pUNO- hMyD88). After 24h of transfection, the cells were either left untreated or treated with 2μM paclitaxel for 48h and caspase-3/7 activity was determined. A significant increase in caspase-3/7 activity was observed in wt cells following paclitaxel treatment. In contrast, no changes in caspase-3/7 activity were observed in cells transfected with pUNO-hMyD88. These findings show that MyD88 expression contributes to paclitaxel chemo-resistance.
Applicants also determined whether the expression of MyD88 confers resistance that is specific to paclitaxel. Wildtype (wt) A2780 EOC cells (carboplatin sensitive) and A2780 EOC cells transfected with MyD88 were treated with 100 μg/ml carboplatin or 2μM paclitaxel for 48 hours. Apoptosis was determined by measuring caspase-3/7 activity. Wildtype A2780 cells showed significant increase in caspase-3/7 activity following treatment with carboplatin or paclitaxel. Upon the introduction of MyD88, the cells retained its sensitivity to carboplatin, but not paclitaxel. These results are evidence that the protective effect of MyD88 is specific for paclitaxel-induced apoptosis.
Example 9: TLR-4 ligation by paclitaxel induces the expression of anti- apoptotic proteins.
TLR-4 ligation through NF-κB and inflammatory molecules promotes cell survival by the induction of the expression of anti-apoptotic proteins (24, 25). The expression of X-linked inhibitor of apoptosis (XIAP), a major inhibitor of capase-3 and 9, and the expression of Akt has been associated with tumor growth and chemoresistance in ovarian cancer cells (26-28). Therefore, Applicants evaluated the expression of XIAP and phosphorylated Akt (pAkt) following paclitaxel treatment in MyD88+ cells. A significant increase in pAkt was observed 4h after treatment with paclitaxel. Similarly, XIAP levels increased 2h post-treatment. In contrast, no change on the levels of total Akt was observed.
Example 10: Correlation between MyD88 expression in ovarian cancer tissues and patients' progression free survival.
In order to determine whether the in vitro findings have clinical relevance, Applicants performed a pilot retrospective study to analyze MyD88 expression in tissues obtained from patients with stage IIIC ovarian cancer (n=12) and determine whether there is a correlation between MyD88 expression and progression free survival. Tissue collection was performed prior to chemotherapy and the expression of MyD88 in ovarian cancer tissues was determined by western blot analysis. All patients received six cycles of paclitaxel/carboplatin regimen after surgery. The mean time to recurrence for patients with tumors that expressed MyD88 was 23 months (n=5). However, the mean time to recurrence for patients with tumors that did not express MyD88 was 42 months (n=7). Patients whose tumors did not express MyD88 had a statistically significant improved progression free interval compared to patients whose tumors expressed MyD88 (p=0.03).
DISCUSSION
Substantial evidence indicates that bacterial- and viral-induced inflammatory processes can mediate tumorigenesis. Described herein is a specific defense mechanism used by the innate immune system, which cancer cells have imitated and adapted to generate proinflammatory cytokines, leading to their own proliferation and survival. Described herein is: 1) the expression of TLR-4 in EOC cells; 2) the induction of tumor growth by TLR-4 ligation in MyD88+ EOC cells; 3) the production of chemokines and cytokines by MyD88+ EOC cells upon TLR-4 ligation; and 4) chemo-resistance to paclitaxel mediated by the expression of MyD88.
It has been observed in animal studies that surgical removal of a primary tumor is often followed by rapid growth of previously dormant metastases and LPS has been suggested to be responsible for this effect (29). Indeed, Balb/c mice receiving a tail vein injection of 4Tl mouse mammary carcinoma cells showed an increase in lung metastases following LPS injection (30). LPS is recognized by TLR-4, which is expressed by the cells of the innate immune system. Following its ligation, it has been shown to induce NFKB activation, cytokines/chemokines production and inflammation (14).
TLRs represent a main receptor pathway, which can induce the expression of pro-inflammatory cytokines (15). TLRs are widely expressed by cells of the immune system and, in response to microbial products or stress factors, initiate an inflammatory process (10). In addition, TLRs have been described in non-immune cells, such as mucosal epithelium and trophoblast cells (20, 31). Similar to immune cells, the ligation of TLRs in non-immune cells results in the expression and secretion of pro-inflammatory cytokines (20). Here, Applicants describe the presence of TLR-4 in all EOC cells tested and the differential expression of MyD88. Applicants showed that in MyD88+ EOC cells, ligation of TLR-4 by LPS enhances cell proliferation and induces the production of chemokines and pro-inflammatory cytokines. Interestingly, unstimulated MyD88+ cells constitutively secrete cytokines/chemokines. This suggests the presence of endogenous ligands of TLRs (e.g., apoptotic bodies, cellular debris from necrotic cells), which can also act through TLR-4 or other TLRs, which are also present in EOC cells (our unpublished data).
One of the main cytokines induced in EOC cells following LPS stimulation is IL-6. In addition, following TLR-4 ligation, these cells secreted the chemokines, MCP-I and GRO-α. It is now well documented that chemokines can dramatically alter the neoplastic process, not only by recruiting leukocytes that will enhance the inflammatory environment, but also by having a direct effect on nearby stromal and neoplastic cells (32). The induction of these chemokines by TLR-4 activation may, therefore, help to recruit inflammatory cells as well as to enhance tumor growth and neoplastic progression. Melanoma is one example in which chemokines have been shown to exert autocrine control over neoplastic proliferation (33, 34). Blocking GRO-α activity on the CXCR2 receptor attenuates melanoma cell proliferation in vitro, whereas over expression of GRO-α and GRO-β enhances tumor colony formation and tumorogenicity in nude mice (34-36). Here, ligation of TLR-4 by LPS induced a significant increase in GRO-α secretion, which may provide the stimuli for the proliferative effect observed on EOC cells following LPS treatment. Furthermore, the presence of pro-inflammatory cytokines and cheniokines has been described as predictors of poor prognosis (37).
As described herein. Applicants have identified a sub-group of epithelial ovarian cancer (EOC) cells that express the protein Myeloid Differentiation Protein 88 (MyD88). MyD88 is an adaptor protein that is required for Toll-like receptor (TLR) signaling, a signaling pathway involved in inflammatory response to bacterial and viral products. EOC cells that express MyD88 constitutively secrete proinflammatory cytokines (IL-6, IL-8, GROα); exhibit high levels of NF-KB; and are resistant to the taxane paclitaxel, which is a known TLR-4 ligand. Upon TLR-4 ligation with either LPS or paclitaxel, these cells secrete higher levels of proinflammatory cytokines and, more importantly, proliferate in culture. Applicants hypothesized that MyD88 is a specific marker for paclitaxel resistance and is required for paclitaxel -induced cell growth in EOC cells. The objective of the work described in this example was to develop an optimized method that can detect MyDS 8 expression in ovarian cancer tumors and thus provide a way for MyD88 as a biomarker for selection of therapy.
Proteomics has emerged as an important tool to study biological processes in both physiological and pathological circumstances. Identification of specific proteins as biomarkers for a disease or condition may be used for diagnosis and/or therapy. Currently however, no optimized method exists that can sensitively detect ovarian cancer biomarkers from tumor tissue prior to initiation of therapy. A limiting factor in the discovery and analysis of potential marker from tumor samples are the "contaminating" signals originating from normal cells, including immune cells, that are infiltrating the tumor. Described herein is a novel approach for the detection of protein expression in a small sample of cancer cells, as exemplified by epithelial ovarian cancer cells dissected by LCM. Furthermore, demonstrated herein is proof that MyD88 expression in LCM-dissected ovarian cancer cells is a marker for paclitaxel resistance.
Applicants observed differential response to LPS by EOC cells, which was due to the expression of the TLR intracellular signaling molecule, MyD88. In the presence of MyD88, treatment with LPS results in the nuclear localization of NF- KB, increase cell proliferation, and cytokine/chemokine production. These results provide preliminary evidence of the involvement of NF-κB in TLR-4-MyD88 signaling pathway in EOC cells. However, the exact downstream signaling pathway after TLR-4-MyD88 activation still remains to be determined.
In the absence of MyD88, treatment with LPS failed to induce NF-κB translocation to the nucleus and had no effect on cellular proliferation nor on cytokine/chemokine production. Interestingly however, the MyD88" A2780 EOC cell line constitutively express nuclear p65 NF-κB, yet does not constitutively express any of the pro-inflammatory cytokines observed in MyD88+ cells. These results suggest that the impact of NF-κB on cancer cells may be cell-type specific and the final output of NF-κB activation, e.g. cytokine production, is the determining factor in tumor progression and differentiation. The significance of this constitutive NF-κB nuclear localization in these cells, which is not related to the production of pro-inflammatory cytokines, is under investigation in our laboratory.
Differential response was also observed in EOC cells in response to paclitaxel. MyD88+ EOC cells responded to paclitaxel in similar manner as they did with LPS: they produced and secreted pro -inflammatory cytokines. In addition, these cells did not undergo apoptosis in response to paclitaxel. This is in contrast to MyD88' cells, which underwent apoptosis and did not secrete cytokine/chemokine in response to paclitaxel.
Studies in mice by Ding et al showed that similar to LPS, treatment with -paclitaxel activates murine macrophages and induces the secretion of inflammatory cytokines, including TNFα, IL-6 and IL-8. This effect of paclitaxel was demonstrated to be both TLR-4 and MyD88 dependent (38) (23). These results are supported by the in vivo observation showing a correlation between expression of MyD88 and progression free survival. Although only a small number of patients was evaluated, the data indicate that the expression of MyD88 correlates with a poor survival.
Applicants also demonstrate that in MyD88+ EOC cells, TLR-4 ligation is able to induce the activation of the Akt survival pathway and enhance the expression of the anti-apoptotic protein XIAP. Both proteins have been shown to be highly expressed in ovarian cancer and are linked to the develop of chemoresistance (17, 27, 39). Therefore, in MyD88+ EOC cells, the pro-apoptotic effect of paclitaxel is overcome by the induction of the anti-apoptotic proteins (pAKT and XIAP) following TLR-4 ligation. Therefore, in these cells, treatment with paclitaxel does not induce apoptosis but induces the secretion of cytokines/chemokines. The correlation between TLR-4-MyD88 signaling and the upregulation of pAkt and XIAP remains to be determined. NF-κB may be a link between these pathways.
During the past twenty years multiple combinations of cytotoxic chemotherapeutics have been evaluated in patients with recurrent ovarian cancer. The work described herein provides new insight into a molecular mechanism that links inflammation and neoplastic development/progression. It suggests that an active TLR-4-MyD88 signaling pathway is a risk factor for developing cancer and a novel target for the development of bio-modulators aimed at reversing chemo- resi stance to paclitaxel.
References for Examples 1-10
1. Beachy PA, Karhadkar SS, Berman DM, Nature 2004;431 (7007):402.
2. Balkwill F, Coussens LM, Nature 2004;431(7007):405-6.
3. Coussens LM, Werb Z5 Nature 2002;420(6917):860-7.
4. Giudice LC3 Kao LC5 Lancet 2004,364(9447): 1789-99.
5. Riman T5 Nilsson S, Persson IR5 Acta Obstet Gynecol Scand 2004;83(9):783-95.
6. Sekizawa A5 Amemiya S, Otsuka J5 et al.5 Med Electron Microsc 2004;37(2):97-100.
7. Robinson SC, Coussens LM5 Adv Cancer Res 2005,93: 159-87.
8. de Visser KE, Coussens LM, Cancer Immunol Immunother 2005,54(1 1):1143-52.
9. Anderson KV5 Bokla L5 Nusslein-Volhard C5 Cell 1985;42(3):791-8.
10. Medzhitov R5 Nat Rev Immunol 2001;l(2):135-45.
11. Medzhitov R5 Janeway CA5 Jr., Cell 1997;91(3):295-8.
12. Takeuchi O5 Akira S, Int Immunopharmacol 2001 ; l(4):625-35.
13. Medzhitov R, Janeway CA5 Jr., Science 2002;296(5566):298-300.
14. Akira S5 J Biol Chem 20035278(40):38105-8.
15. Akira S, Takeda K, Nat Rev Immunol 2004,4(7) :499-511. 16. Behrens BC, Hamilton TC, Masuda H5 et al., Cancer Res 1987;47(2):414-8.
17. Kamsteeg M, Rutherford T5 Sapi E5 et al., Oncogene 2003;22(17):2611-20.
18. Flick MB5 O'Malley D5 Rutherford T5 et al., J Soc Gynecol Inv 2004; 11:252- 9.
19. Alvero AB, Fishman DA, Qumsiyeh MB5 Garg M, Kacinski BM, Sapi E, J Soc Gynecol Investig 2004;l 1(8):553-61.
20. Abrahams VM5 Bole- Aldo P, Kim YM5 et al., J Immunol 2004; 173(7):4286- 96.
21. O'Dwyer PJ, Moyer JD, Suffness M, et al., Cancer Res 1994;54:724-9.
22. Kaplan E, Meier P., J Amm Stat Ass 1958;55:457-81.
23. Byrd-Leifer CA, Block EF, Takeda K, Akira S5 Ding A, Eur J Immunol 2001;31(8):2448-57.
24. Schmitz I5 Kirchhoff S, Krammer PH, Int J Biochem Cell Biol 2000;32(l 1- 12):1123-36.
25. Nakanishi C, Toi M, Nat Rev Cancer 2005;5(4):297-309.
26. Yuan ZQ, Feldman RI, Sussman GE, Coppola D, Nicosia SV, Cheng JQ, J Biol Chem 2003;278(26):23432-40.
27. Dan HC, Sun M, Kaneko S, et al., J Biol Chem 2004;279(7):5405-12.
28. Dan HC, Jiang K, Coppola D, et al., Oncogene 2004;23(3):706-15.
29. Pidgeon GP, Harmey JH, Kay E, Da Costa M, Redmond HP, Bouchier- Hayes DJ, Br J Cancer 1999;81(8):1311-7.
30. Harmey JH, Bucana CD, Lu W, et al, Int J Cancer 2002;101(5):415-22. •
31. Abrahams VM, Mor G, Placenta 2005;26(7):540-7.
32. Vicari AP, Caux C, Cytokine Growth Factor Rev 2002; 13(2): 143-54.
33. Richmond A, Thomas HG, J Cell Physiol 1986;129(3):375-84.
34. Richmond A, Fine R5 Murray D, Lawson DH, Priest JH, J Invest Dermatol 1986;86(3):295-302.
35. Metzner B, Barbisch M, Bachmann F, Czech W, Norgauer J, J Invest Dermatol 1996;107(4):597-602.
' 36. Norgauer J5 Metzner B, Schraufstatter I, J Immunol 1996; 156(3): 1132-37.
37. McMillan DC5 Canna K, McArdle CS, Br J Surg 2003;90(2):215-9.
38. Ding AH, Porteu F, Sanchez E, Nathan CF, Science 1990;248(4953):370-2. 39. Sapi E, Chen W5 O'Malley D, et al, Anti-Cancer Drugs 2004;14:567-78.
MATERIALS AND METHODS
The following materials and methods were used in Examples 11-13.
Patients and samples preparation
Ovarian cancer tissue samples were collected from advanced stage ovarian cancer patients at the time of surgery. All patients signed consent forms and the use of patient samples was approved under Yale University's Human Investigations Committee (HIC #10425).
Tissues were prepared in small aliquots and snap-frozen in liquid N2. AU samples were stored in cryovials at -800C until future use.
Antibodies and reagents for immunocytochemistry (IHC)
Albumin from bovine serum / BSA was purchased from Sigma Aldrich, St. Louis, MO.
The primary antibody was Dako OV-TL mouse monoclonal cytokeratin-7 from Dakocytomation, Carpinteria, CA. Primary antibody dilution was 1 :150 in 1% BSA mixed in wash buffer. The secondary antibody used was biotinylated anti- mouse IgG (H+L) made in horse from Vector Laboratories, Burlingame, CA. Dilution was 1 :200 in 1% B S A/wash buffer. As detection reagent we used Streptavidin-HRP Conjugate from Zymed, San Francisco, CA, in a 1 :300 dilution.
Antibodies and reagents for Western blot analysis
Primary antibodies used for Western blot analysis were rabbit polyclonal anti-human and mouse antibody to MyD88 (dilution 1:1000 in PBS-T/1% FFPM) from eBioscience (San Diego, CA), mouse monoclonal anti-human antibodies to FasL/CD95L (dilution 1 :10,000 in PBS-T/1% FFPM) from BD Biosciences (San Jose, CA), mouse monoclonal anti-human antibodies to hIAL/XIAP (dilution 1:10,000 in PBS-T/1% FFPM) from BD Biosciences PharMingen (San Diego, CA). Secondary peroxidase-conjugated antibodies, anti-rabbit IgG (H+L) made in goat and anti-mouse IgG (H+L) made in horse, were purchased from Vector Laboratories (Burlingame, CA). Slide preparation
In preparation for Laser capture microdissection a 5x5 mm tissue sample was embedded in OCT at -200C. Eight-micron sections were cut with the microtome and collected on Leica glass-foiled PEN-membrane slides. The slides were fixed in 95% ethanol for 5 minutes and stained with H&E for 30 seconds at RT, followed by washing in ddH2O for 30 seconds. Dehydration is required for successful Laser microdissection and this was accomplished by immersing the slides in 95% ethanol and then in Histosolve, a xylene substitute (Shandon, Inc., Pittsburgh, PA), 15 minutes each. The slides were air dried at RT for 15 minutes. Following dehydration, to prevent protein degradation, the samples were either used immediately for microdissection or they were stored in air-tight plastic wrapping at - 200C. Immunohistochemistry for Laser microdissection
Eight-micron sections were cut with the microtome, collected on Leica glass- foiled PEN-membrane slides and fixed in 95% ethanol for 5 minutes as described above. The slides were then placed in a staining dish with 0.1% H2O2 in 0.1 M PB for 10 minutes to quench for endogenous peroxidase activity and then washed in wash buffer 3 times. Wash buffer was prepared by adding 0.01% Triton X-100 (Sigma Aldrich, St. Louis, MO) to 0.1 M PB. The area around the tissue sections was scored with a Pap pen to limit the amount of antibodies and reagents used. All steps occurred at room temperature with the slides placed in a moisture chamber to keep the tissue from drying out during the procedure. To block for non-specific background, 100-200 μl of 3% BSA made in wash buffer were added to the circumscribed areas and incubated for 20 minutes in the moisture chamber. Next, the primary antibody was incubated for 2 hours followed by washing with wash buffer (each wash is 2-3 minutes x3), the specimens were incubated with Biotin labeled secondary antibody for 30 minutes. After this incubation, the slides were washed once with wash buffer then twice with 0. IM Tris (pH 7.5). The tissue was incubated with Streptavidin-HRP conjugate for 30 minutes and then washed with 0.1 M Tris 3 times. The color was developed with DAB (2.5 mg DAB in 5 ml 0.1 M Tris). Twenty five μl of 0.03% H2O2 were added to the chromogen just before use. At the end of the procedure the slides were washed with double distilled water and counterstained with Mayer's hematoxylin (Sigma Aldrich, St. Louis, MO). The specimens were dehydrated and then stored as described above. Laser Microdissection for Western Blot Analysis
The slides were placed inverted into the holder of a Leica AS LMD Laser Microdissection System with the side containing the tissue facing down (non-contact method). Tumor cells were selected according to their morphology for H&E stained samples or according to their immunoreactivity for CK-7 stained samples. Dissected cells were collected in the cup of 0.5ml Eppendorf tubes containing 35 μl sample buffer (2.5% SDS, 10% glycerol, 5% β-mercapto-ethanol, 0.15M Tris (pH = 6.8) and 0.01% bromophenol blue). After a short centrifugation, lysis of the cells was achieved by submitting the samples to 5 cycles of freezing (liquid N2) and thawing (95 0C) followed by boiling for 5 minutes, centrifuged again for 2 minutes and then stored at -8O0C until further use..
All procedures were carried out in a timely manner to minimize the activity of proteases. For H&E stained tissues, the interval from tissue frozen section cutting to cell dissection and lysis was under 1 hour while immunostained (CK-7 or CD45) stained specimens required approximately 4 hours.
Western Blot Analysis
Proteins were resolved under reducing conditions on 12% SDS-PAGE gels and then transferred onto polyvinylidene difluoride paper (NEN Life Sciences, Boston, MA) (5). Membranes were blocked at room temperature for 1 hour with 5% FFPM in PBS/0.05%Tween 20 (PBS-T). After three washes for 10 minutes, each with PBS-T, membranes were incubated overnight at 40C with the primary antibody in PBS-T/1% FFPM. After incubation with the primary antibody, membranes were washed three times with PBS-T and then incubated at room temperature with the appropriate secondary antibody conjugated to peroxidase (Vector Laboratories) in PBS-T/1% FFPM. After three washes for 10 minutes each with PBS-T and three washes for 10 minutes each with distilled water, the peroxidase-conjugated antibody was detected by enhanced chemoluminiscence (PerkinElmer Life Sciences, Boston, MA). The signal intensity was analyzed using a Kodak digital imaging analysis system (Kodak Image Station 4000MM) and Kodak Molecular Imaging Software (Scientific Imaging Kodak Company). Example 11 Protein detection in isolated cells
Results demonstrated that protein expression can be detected by Western blot analysis in microdissected cells obtained from ovarian tumor samples that were snap-frozen in liquid N2. Increasing number of cells were microdissected with a maximum of 8,000 cells and a minimum of 100 cells from 8 μm ovarian cancer sections. These were analyzed for the expression of MyD88 and other cancer- related proteins, such as XIAP and FasL. A strong signal for MyD88 expression was observed in samples containing 8,000 and 5,000 microdissected cells; no signal was detected for samples containing fewer than 1 ,000 cells.
In order to determine whether the sensitivity of the assay remains similar for different proteins, Applicants evaluated the expression of XIAP and FasL using the same number of cells as for MyD88. In contrast to MyD88, XIAP and FasL expression is detected in samples containing as few as 1000 and 100 cells, respectively.
Whether sample thickness affects the signal detected by Western blot was also evaluated. Although increasing the section thickness increased the amount of protein, it affected cutting time and rendered dissection extremely difficult. Therefore, the method was standardized to 8 μm sections. This allows a quick dissection and provides enough protein to be detected by Western blot.
As a result, Applicants demonstrated that fresh-frozen tissue is a suitable material for LCM. They showed that proteins involved in the apoptolic cascade and chemoresistance of EOC cells can be accurately detected by, for example, snap freezing tumor specimens in liquid nitrogen, and staining specimens (e.g., 8 μm thick specimens to facilitate selection of pure tumor cells; identifying target cells, based on their immunostaining and collecting target cells (e.g., with the LCM system); and Iy sing collected target cells, followed by analysis for protein content/makeup (e.g., by Western blot or Luminex multiplex analysis).
Example 12 MyD88 expression is cell-type specific Tumors are heterogeneous tissues containing normal supporting cells, immune cells and cancer cells. Immune cells infiltrate the stroma and may "contaminate" the protein profile of the tumor sample. Indeed, when Applicants analyzed MyD88 expression in H&E stained samples without using appropriate markers for cell type identification, they found that MyD88 was expressed in all samples. The explanation for these results is that the immune cells present in the tumor tissue and surrounding the cancer cells express MyD88. In an H&E stained sample, the selection of specific cells based on their morphology alone is not sufficient to identify intra-tumoral infiltrated cells. An advantage of the LCM is that it affords the possibility of selecting a specific group of cells based on the expression of cellular markers. Therefore, Applicants' next objective was to determine whether immunohistochemistry staining affects the detection of proteins in microdissected samples. Thus, they compared the sensit ) ivity of protein detection of cells obtained
/ from H&E stained samples with cells obtained from immuno-stained samples. Tumor tissues were immuno-stained with antibodies for cytokeratin 7 (epithelial marker) or CD45 (pan-leucocyte marker). Specific staining for cytokeratin 7 and CD45 was observed in these frozen sections. Comparison of MyD88 and XIAP expression in cells obtained from these two preparations, showed no difference in the signal intensity between the two groups, suggesting that the process of IHC does not affect the samples' integrity. After establishing that IHC does not affect protein detection, Applicants evaluated MyD88 expression in ovarian cancer samples after staining for CD45 (panleukocyte antigen). Assessment of only cancer cells (CD45 negative cells), avoiding immune cells (CD45 positive cells), showed that MyD88 was differentially expressed in tumors from different patients with identical pathological diagnoses.
Example 13 Clinical correlation between MyD88 expression and Survival/Progression Free Interval
Applicants evaluated 20 tumor samples from patients with epithelial ovarian cancer. Eleven out of twenty tumors were MyD88 positive and nine tumors were MyD88 negative.
From the MyD88 positive tumors, 7/11 were obtained at the time of initial surgery from patients who received neoadjuvant chemotherapy with carboplatin and paclitaxel. These patients had persistent or progression of disease during neoadjuvant chemotherapy. Even after neoadjuvant chemotherapy and optimal tumor debulking, these patients failed postoperative chemotherapy. Their progression-free interval was zero and four patients died of disease in less than 1 year. In this group, mean survival was 16 months.
From the MyD88 negative tumor samples, 5/7 were obtained at the time of initial surgery from patients with poorly differentiated serous papillary adenocarcinomas. Three patients were stage IIIC, one was stage IIIB, and one was stage IIC. In this group all patients were treated with carboplatin and paclitaxel and the mean progression-free interval was 35 months. The mean overall survival in this group was 45 months and three patients are alive at the time of this report. Four of eleven MyD88 positive tumors were obtained from patients with recurrent disease who underwent optimal secondary cytoreductive surgery. All four patients (two with recurrent stage IC, one with recurrent stage HA, and one with recurrent stage IIC) received carboplatin and paclitaxel. Their mean progression-free interval after completion of chemotherapy was 3 months.
Four MyD88 negative tumor samples were obtained from patients with recurrent disease at the time of the secondary cytoreduction. Postoperatively, these patients were treated with paclitaxel and carboplatin combination chemotherapy. One patient with stage IIIC recurred 33 months after primary treatment and one patient with stage IVA recurred 20 months after primary treatment. Mean progression-free interval in this group was 14 months.
A statistical significant correlation between assay prediction of response and progression-free interval (PFI) was observed in all these cases. Furthermore, patients who had MyD88 negative tumors responded better to treatment and had a significantly longer survival when compared with the patients who had MyD88 positive tumors (Figures 1 and 2). References for Examples 11-13
1. Schwartz PE. Current diagnosis and treatment modalities for ovarian cancer. Cancer Treat Res 2002;107:99-l 18.
2. Kelly MG5 Alvero AB, Chen R, Silasi DA, Abrahams VM, Chan S, et al. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res 2006;66(7):3859-3868.
3. Ding Y, Xu L5 Chen S, Jovanovic BD, Helenowski IB, Kelly DL, et al Characterization of a method for profiling gene expression in cells recovered from intact human prostate tissue using RNA linear amplification. Prostate Cancer Prostatic Dis 2006.
4. Matsuzaki S, Canis M, Pouly JL, Botchorishvili R, Dechelotte PJ, Mage G. Differential expression of genes in eutopic and ectopic endometrium from patients with ovarian endometriosis. Fertil Steril 2006;86(3):548-553.
5. Alvero AB, O'Malley D, Brown D, Kelly G, Garg M3 Chen W, et al. Molecular mechanism of phenoxodiol-induced apoptosis in ovarian carcinoma cells. Cancer 2006;106(3):599-608.
6. Asselin E, Mills GB, Tsang BK. XIAP regulates Akt activity and caspase-3- dependent cleavage during cisplatin-induced apoptosis in human ovarian epithelial cancer cells. Cancer Res 2001 ;61 (5): 1862-1868.
7. Holcik M, Gibson H, Korneluk RG. XIAP: apoptotic brake and promising therapeutic target. Apoptosis 2001 ;6(4):253-261.
8. Sapi E, Chen W, O'Malley D, Hao X, Dwipoyono B, Garg M, et al. Resistance of Ovarian Cancer Cells to Docetaxel is XIAP Dependent and Reversible by Phenoxodiol. Anti-Cancer Drugs 2004; 14:567-578.
9. Gutierrez L, Eliza M, Niven-Fairchild T, Naftolin F, Mor G. The Fas/Fas- Ligand system: A mechanism for immune evasion in human breast carcinomas. Breast Cancer Research and Treatment 1999;54(3):245-253.
10. Abrahams V, Straszewski S, Kamsteeg M, Hanczaruk B, Schwartz P, Rutherford T, et al. Epithelial Ovarian Cancer secrete funcitonal Fas Ligand. Cancer Res 2003 ;63 =5573-5581.

Claims

1. A method of assessing responsiveness of a cancer cell to a plant alkaloid, comprising determining whether the cancer cell expresses MyD88 (MyDSS+) or does not express MyD88 (MyD88")5 wherein if the cancer cell is MyD88+, it is resistant to the plant alkaloid and if the cancer cell is MyD88", it is sensitive to the plant alkaloid.
2. The method of claim 1 , wherein the plant alkaloid is a taxane.
3. The method of claim 1, wherein the plant alkaloid is paclitaxel or docetaxel.
4. The method of claim 2, wherein the taxane is paclitaxel or docetaxel
5. The method of any one of claims 1 to 4, wherein the cancer cell is an ovarian cancer cell, a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell
6. The method of any one of claims 1 to 5, wherein the cancer cell is an epithelial ovarian cancer cell.
7. The method of any one of claims 1 to 7, wherein the cancer cell is a human cancer cell.
8. A method of assessing sensitivity or resistance of a cancer cell to chemotherapy with a plant alkaloid, comprising assaying expression of MyD88 in the cancer cell, wherein the cancer cell is sensitive to chemotherapy with the plant alkaloid if the cancer cell does not express MyD88 and the cancer cell is resistant to chemotherapy with the plant alkaloid if the cancer cell expresses MyD88.
9. The method of claim 8, wherein the plant alkaloid is a taxane.
10. The method of claim 8, wherein the plant alkaloid is paclitaxel or docetaxel.
11. The method of claim 9, wherein the taxane is paclitaxel or docetaxel.
1149836.1 39
12. The method of any one of claims 8 to 11, wherein the cancer cell is an ovarian cancer cell, a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell.
13. The method of any one of claims 8 to 12, wherein the cancer cell is an epithelial ovarian cancer cell.
14. The method of any one of claims 8 to 13, wherein the cancer cell is a human cancer cell.
15. The method of any one of claims 1 to 7, wherein determining whether the cancer cell expresses MyD88 or does not express MyD88 is carried out by detecting the presence or absence of MyD88 in the cancer cell, detecting the quantity of MyD88 in the cancer cell or detecting nucleic acid that encodes MyD88 in the cancer cell.
16. The method of any one of claims 8 to 14, wherein assaying expression of MyD88 in the cancer cell is carried out by detecting the presence or absence of MyD88 in the cancer cell, detecting the quantity of MyD88 in the cancer cell or detecting nucleic acid that encodes MyD88 in the cancer cell.
17. The method of claim 15 or 16, wherein MyD88 is detected by Western blot analysis.
18. A method of inhibiting proliferation of a cancer cell, comprising contacting the cancer cell with a compound which inhibits signaling through the TLR4:MyD88 pathway.
19. The method of claim 18, wherein the compound is a TLR4 antagonist.
20. The method of claim 19, wherein the compound inhibits activity or expression of TLR-4.
21. The method of claim 19, wherein the TLR4 antagonist is an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA.
22. The method of claim 18, wherein the compound is a MyD88 antagonist.
23. The method of claim 22, wherein the compound inhibits activity or expression of MyD88.
24. The method of claim 22, wherein the MyD88 antagonist is selected from an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
25. The method of claim 18, further comprising assessing signaling through the TLR4:MyD88 pathway after the cancer cell is contacted with the compound, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF-κB.
26. The method of claim 18, further comprising assessing signaling through the TLR4:3VIyD88 pathway after the cancer cell is contacted with the compound, wherein signaling through the TLR4:MyD88 pathway is measured by determining the presence or absence of a pro-inflammatory cytokine or the level of a pro- inflammatory cytokine.
27. The method of claim 26, wherein the pro-inflammatory cytokine is GRO-α, MCP-I or IL-6.
28. The method of any one of claims 18 to 27, wherein the cancer cell is an ovarian cancer cell, a breast cancer cell, a head cancer cell, a neck cancer cell, a prostate cancer cell, a non-small cell lung cancer cell, a small cell lung cancer cell, a bladder cancer cell, a stomach cancer cell, or a Kaposi's sarcoma cell
29. The method of any one of claims 18 to 28, wherein the cancer cell is an epithelial ovarian cancer cell.
30. The method of any of claims 18 to 29, wherein the cancer cell is a human cancer cell.
31. A method of treating or preventing cancer in an individual, comprising administering to the individual an effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway.
32. The method of claim 31, wherein the compound is a TLR4 antagonist.
33. The method of claim 32, wherein the compound inhibits activity or expression of TLR-4.
34. The method of claim 32, wherein the TLR4 antagonist is an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound, an antisense nucleic acid, or an siRNA.
35. The method of claim 31, wherein the compound is a MyD88 antagonist.
36. The method of claim 35, wherein the compound inhibits activity or expression of MyD88.
37. The method of claim 35, wherein the MyD88 antagonist is selected from an antibody against MyD88, a mutated form of MyD88, a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
38. The method of claim 31 , further comprising assessing signaling through the TLR4:MyD88 pathway after the effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway is administered to the individual, wherein signaling through the TLR4:MyD88 pathway is measured by activation status of NF-κB.
39. The method of claim 31, further comprising assessing signaling through the TLR4:MyD88 pathway after the effective amount of a compound that inhibits signaling through the TLR4:MyD88 pathway is administered to the individual, wherein signaling through the TLR4:MyD88 pathway is measured by determining the presence or absence of a pro-inflammatory cytokine or the level of a proinflammatory cytokine.
40. The method of claim 39, wherein the pro-inflammatory cytokine is GRO-α, MCP-I or IL-6.
41. The method of any one of claims 31 to 40, wherein the cancer is ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
42. The method of any one of claims 31 to 40, wherein the cancer is epithelial ovarian cancer.
43. The method of any one of claims 31 to 42, wherein the cancer cell is a human cancer cell.
44. The method of any one of claims 31 to 43, wherein the cancer is characterized by cancer cells that do not express MyD88 (MyD88" cells).
45. The method of claim 44, wherein the individual is a human.
46. An isolated therapeutic compound which specifically inhibits signaling through the TLR4:MyD88 pathway.
47. The therapeutic compound of claim 46, wherein the compound is a TLR4 antagonist.
48. The therapeutic compound of claim 46, wherein the compound inhibits activity or expression of TLR4.
49. The therapeutic compound of claim 47, wherein the TLR4 antagonist is selected from an antibody against TLR4, a mutated form of TLR4, a mimic of TLR4, a small molecule compound an anti sense nucleic acid, and an siRNA
50. The therapeutic compound of claim 46, wherein the compound is a MyD88 antagonist.
51. The therapeutic compound of claim 50, wherein the compound inhibits activity or expression of MyD88.
52. The therapeutic compound of claim 50, wherein the MyD88 antagonist is selected from an antibody against MyD88, a mutated form or a mimic of MyD88, a small molecule compound, an antisense nucleic acid, and an siRNA.
53. A method for monitoring drug treatment of an individual who has cancer, comprising:
a) administering a drug to the individual; and b) assaying the expression level of MyD88, wherein the expression level of MyD88 is an indicator of the disease status of the individual.
54. The method of claim 53, wherein the cancer is ovarian cancer.
55. The method of claim 53 or claim 54, wherein the individual is a human.
56. The method of any one of claims 53 to 55, wherein the cancer does not express MyD88 (MyD88").
57. The method of claim 53, wherein the cancer is ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
58. A method of enhancing chemo-sensitivity in an individual who has cancer, comprising administering to an individual in need thereof a therapeutically effective amount of a compound which inhibits signaling through the TLR4:MyD88 pathway.
59. The method of claim 58, wherein the individual has ovarian cancer, breast cancer, head and/or neck cancer, prostate cancer, non-small cell lung cancer, small cell lung cancer, bladder cancer, stomach cancer, or Kaposi's sarcoma.
60. A method of diagnosing or aiding in diagnosing cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD88 which is greater than a reference expression level is indicative of tumor tissue or precancerous tissue in the individual.
61. The method of claim 60, wherein the cancer is ovarian cancer.
62. A method for screening for genetic susceptibility to cancer in an individual, comprising assaying the expression level of MyD88 in a tissue from the individual, wherein an expression level of MyD 88 which is greater than a reference expression level is indicative of genetic susceptibility to cancer in the individual.
63. The method of claim 62, wherein the cancer is ovarian cancer.
64. The method of claim 18, wherein the cancer cell is in a human.
PCT/US2007/004707 2006-02-23 2007-02-23 Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway WO2007100650A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/224,244 US7985538B2 (en) 2006-02-23 2007-02-23 Drug resistance and methods of reversing
EP07751467A EP1996935B1 (en) 2006-02-23 2007-02-23 Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway
AT07751467T ATE524732T1 (en) 2006-02-23 2007-02-23 DRUG RESISTANCE TO PLANT ALKALOIDS IS BASED ON THE MYD88 STATUS IN A CELL AND METHOD FOR INHIBITING SIGNALING THROUGH THE TLR-4:MYD88 SIGNALING PATHWAY

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US77630406P 2006-02-23 2006-02-23
US60/776,304 2006-02-23
US78816806P 2006-03-31 2006-03-31
US60/788,168 2006-03-31

Publications (2)

Publication Number Publication Date
WO2007100650A2 true WO2007100650A2 (en) 2007-09-07
WO2007100650A3 WO2007100650A3 (en) 2008-04-03

Family

ID=38349578

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/004707 WO2007100650A2 (en) 2006-02-23 2007-02-23 Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway

Country Status (4)

Country Link
US (1) US7985538B2 (en)
EP (1) EP1996935B1 (en)
AT (1) ATE524732T1 (en)
WO (1) WO2007100650A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010020590A1 (en) * 2008-08-20 2010-02-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting the response to anti-cancer treatment with an agonist of tlr7 or an agonist of tlr8
EP2733491A1 (en) * 2006-03-27 2014-05-21 Isis Innovation Limited Diagnostic method
US8901171B2 (en) 2010-01-27 2014-12-02 Takeda Pharmaceutical Company Limited Compounds for suppressing a peripheral nerve disorder induced by an anti-cancer agent
JP2015108639A (en) * 2009-05-22 2015-06-11 ザ・ユナイテッド・ステイツ・オブ・アメリカ・アズ・リプリゼンティド・バイ・ザ・セクレタリー・フォー・ザ・デパートメント・オブ・ヘルス・アンド・ヒューマン・サービシズ Akt phosphorylation at ser473 as indicator for taxane-based chemotherapy
WO2019055597A1 (en) * 2017-09-13 2019-03-21 National Tuberous Sclerosis Association, Inc. Methods and compositions for the treatment of tsc
CN111643677A (en) * 2020-05-26 2020-09-11 四川省肿瘤医院 Nanoliposome for encapsulating TLR4/MyD88 signal channel antagonist and preparation method and application thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115715802A (en) * 2022-09-09 2023-02-28 上海市普陀区中心医院 Application of MyD88 inhibitor in preparation of medicine for treating breast cancer and reversing paclitaxel resistance of breast cancer
CN117418013A (en) * 2023-12-19 2024-01-19 四川省肿瘤医院 Primer, method and application for detecting methylation level of ovarian cancer cell MyD88 gene 5' URR CpG island

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1697718A4 (en) 2003-11-26 2007-11-28 Univ Yale Apoptosis-based evaluation of chemosensitivity in cancer patients
AU2006301230A1 (en) 2005-10-12 2007-04-19 Cancer Research Technology Ltd. Methods and compositions for treating immune disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SCHWARTZ PE, CANCER TREAT RES, vol. 107, 2002, pages 79 - 118

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2733491A1 (en) * 2006-03-27 2014-05-21 Isis Innovation Limited Diagnostic method
US8784806B2 (en) 2006-03-27 2014-07-22 Isis Innovation Limited Screening method
US9347103B2 (en) 2006-03-27 2016-05-24 Isis Innovation Limited Screening method
US10012652B2 (en) 2006-03-27 2018-07-03 Oxford University Innovation Limited Screening method
WO2010020590A1 (en) * 2008-08-20 2010-02-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting the response to anti-cancer treatment with an agonist of tlr7 or an agonist of tlr8
US8772243B2 (en) 2008-08-20 2014-07-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for predicting the response to anti-cancer treatment with an agonist of TLR7 or an agonist of TLR8
JP2015108639A (en) * 2009-05-22 2015-06-11 ザ・ユナイテッド・ステイツ・オブ・アメリカ・アズ・リプリゼンティド・バイ・ザ・セクレタリー・フォー・ザ・デパートメント・オブ・ヘルス・アンド・ヒューマン・サービシズ Akt phosphorylation at ser473 as indicator for taxane-based chemotherapy
US8901171B2 (en) 2010-01-27 2014-12-02 Takeda Pharmaceutical Company Limited Compounds for suppressing a peripheral nerve disorder induced by an anti-cancer agent
WO2019055597A1 (en) * 2017-09-13 2019-03-21 National Tuberous Sclerosis Association, Inc. Methods and compositions for the treatment of tsc
CN111643677A (en) * 2020-05-26 2020-09-11 四川省肿瘤医院 Nanoliposome for encapsulating TLR4/MyD88 signal channel antagonist and preparation method and application thereof
CN111643677B (en) * 2020-05-26 2023-06-27 四川省肿瘤医院 Nanometer liposome for encapsulating TLR4/MyD88 signal pathway antagonist and preparation method and application thereof

Also Published As

Publication number Publication date
WO2007100650A3 (en) 2008-04-03
EP1996935B1 (en) 2011-09-14
US7985538B2 (en) 2011-07-26
US20090220427A1 (en) 2009-09-03
EP1996935A2 (en) 2008-12-03
ATE524732T1 (en) 2011-09-15

Similar Documents

Publication Publication Date Title
EP1996935B1 (en) Drug resistance to plant alkaloids based upon myd88 status in a cell and methods of inhibiting signaling through the tlr-4:myd88 pathway
Kelly et al. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer
JP6234967B2 (en) Cancer markers and therapeutic targets
Wei et al. Dishevelled family proteins are expressed in non-small cell lung cancer and function differentially on tumor progression
Silasi et al. Cancer issue: MyD88 predicts chemoresistance to paclitaxel in epithelial ovarian cancer
Radosavljevic et al. Interleukin-17 may be a valuable serum tumor marker in patients with colorectal carcinoma
Atasoy et al. Fas-mediated pathway and apoptosis in normal, hyperplastic, and neoplastic endometrium
Enjoji et al. The tumor-associated antigen, RCAS1, can be expressed in immune-mediated diseases as well as in carcinomas of biliary tract
Yu et al. CIP2A is overexpressed in human endometrioid adenocarcinoma and regulates cell proliferation, invasion and apoptosis
WO2012100573A1 (en) New molecular marker cuedc2 protein for prognostic determination of breast cancer endocrinology therapy
Zhang et al. EFEMP1 binds to STEAP1 to promote osteosarcoma proliferation and invasion via the Wnt/β-catenin and TGF-β/Smad2/3 signal pathways
Leung et al. Expressions of c-erbB-2, epidermal growth factor receptor and pan-ras proto-oncogenes in adenocarcinoma of the cervix: correlation with clinical prognosis
CN114395625B (en) Application of COPA in preparation of cervical cancer diagnosis biomarker and/or development of cervical cancer drug
Zhu et al. HIP1R acts as a tumor suppressor in gastric cancer by promoting cancer cell apoptosis and inhibiting migration and invasion through modulating Akt
KR101988120B1 (en) Diagnosis of gastric cancer using gastrokine 1 protein within blood
EP2155900A1 (en) Deletion bearing bard1 isoforms and use thereof
KR102401005B1 (en) A composition for preventing or treating high-risk endometriosis
JP2019158613A (en) Detection of placental site trophoblastic tumor (pstt) using laeverin
Meert et al. Angiogenesis in preinvasive, early invasive bronchial lesions and micropapillomatosis and correlation with EGFR expression
Abdel-hamied et al. The Programmed Death-1 Receptor, Programmed Death-1 Ligand (PD-1/PD-L1) and apoptosis in breast cancer patients: A potential Mechanism of immune escape
Belal et al. Assessment of immunhistochemical expression of CD44 and osteopontin in colorectal carcinoma
KR20210127489A (en) Biomarker for the diagnosis of PD-L1 inhibitor and CCL-2 inhibitor resistant cancer and use thereof
CN114622017A (en) Application of ISG15 and ISG15 induced macrophages and secretion thereof in tumor treatment
WO2004018510A1 (en) Marker for cancer
Nagarsheth et al. 0098: Feasibility of Bloodless Surgery On a Gynecolo-Giconcology Service

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007751467

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12224244

Country of ref document: US